WO2019075200A1 - Transgenic selection methods and compositions - Google Patents

Transgenic selection methods and compositions Download PDF

Info

Publication number
WO2019075200A1
WO2019075200A1 PCT/US2018/055412 US2018055412W WO2019075200A1 WO 2019075200 A1 WO2019075200 A1 WO 2019075200A1 US 2018055412 W US2018055412 W US 2018055412W WO 2019075200 A1 WO2019075200 A1 WO 2019075200A1
Authority
WO
WIPO (PCT)
Prior art keywords
terminal fragment
intein
protein
nucleotide sequence
sequence encoding
Prior art date
Application number
PCT/US2018/055412
Other languages
French (fr)
Inventor
Albert Cheng
Nathaniel JILLETTE
Menghan DU
Original Assignee
The Jackson Laboratory
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Jackson Laboratory filed Critical The Jackson Laboratory
Priority to CA3079017A priority Critical patent/CA3079017A1/en
Priority to US16/755,065 priority patent/US20200263197A1/en
Priority to CN201880078542.7A priority patent/CN111511759A/en
Priority to JP2020520468A priority patent/JP7394752B2/en
Priority to AU2018347421A priority patent/AU2018347421A1/en
Priority to EP18867279.4A priority patent/EP3694869A4/en
Priority to KR1020207013411A priority patent/KR20200064129A/en
Publication of WO2019075200A1 publication Critical patent/WO2019075200A1/en
Priority to JP2023200808A priority patent/JP2024015079A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/1029Acyltransferases (2.3) transferring groups other than amino-acyl groups (2.3.1)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/61Fusion polypeptide containing an enzyme fusion for detection (lacZ, luciferase)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/90Fusion polypeptide containing a motif for post-translational modification
    • C07K2319/92Fusion polypeptide containing a motif for post-translational modification containing an intein ("protein splicing")domain
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3517Marker; Tag
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3519Fusion with another nucleic acid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • Antibiotic resistance genes encoding antibiotic resistance proteins
  • Antibiotic resistance genes/antibiotics available for use in eukaryotic cells include /zygfi/Hygromycin, neol Geneticin ® /G418, /?ac/Puromycin, Sh Wa/Phleomycin Dl (Zeocin TM ), and fed/Blasticidin.
  • Fluorescent proteins, such as green fluorescent protein (GFP) provide another means of cell selection, for example, via fluorescent- activated cell sorting (FACS) techniques or fluorescent microscopy.
  • compositions and kits useful for the production and/or identification of, for example, cells and/or organisms harboring two or more transgenes (e.g., double-transgenics, triple-transgenics, etc.).
  • the compositions and kits may be used for the production and/or identification of cells and/or organisms harboring two, three, or four transgenes.
  • This technology is based, at least in part, on a protein splicing mechanism initiated by an intein auto-processing domain, which facilitates the joining (conjugation) specifically in multi- transgenic cells of multiple (e.g., two, three, or four) separate selectable marker protein fragments (double-transgenic cells, triple-transgenic eels, or quadruple-transgenic cells).
  • Joining of the two, three, four, or more separate selectable marker protein fragments in the multi-transgenic cells produces a full-length selectable marker protein that confers, for example, antibiotic resistance (an antibiotic resistance protein) or is capable of fluorescence under an appropriate wavelength of light (fluorescent protein).
  • Cells expressing a full-length antibiotic resistance gene survive in the presence of the corresponding antibiotic and thus are selected as multi-transgenic (e.g., double- transgenic, triple-transgenic, or quadruple-transgenic) cells.
  • multi-transgenic e.g., double- transgenic, triple-transgenic, or quadruple-transgenic
  • cells expressing a full-length functioning fluorescent protein fluoresce under the appropriate wavelength of light and thus are selected as multi-transgenic (e.g., double-transgenic, triple-transgenic, or quadruple-transgenic) cells.
  • the present disclosure provides, in some embodiments, methods comprising delivering to a composition comprising eukaryotic cells two or more vectors, wherein each vector comprises (i) a nucleotide sequence encoding a selectable marker protein fragment linked to an N-terminal intein protein fragment and/or a C-terminal intein protein fragment and (ii) a nucleotide sequence encoding a molecule of interest, wherein the intein protein fragments, when joined in frame to form full-length function proteins, catalyze joining of the selectable marker protein fragments to produce a full-length selectable marker protein.
  • methods herein comprising delivering to a composition comprising eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding a first selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) upstream from a nucleotide sequence encoding an N-terminal intein protein fragment and (ii) a nucleotide sequence encoding a first molecule, and (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) and (ii) a nucleotide sequence encoding a second molecule, wherein the N- terminal intein protein fragment and the C -terminal intein protein fragment catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length selectable marker protein.
  • the two vectors When the two vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, and some cells will take up both vectors. Only those cells that take up both vectors are capable of expressing a full-length functioning selectable marker protein, thus only those cells are selected as double-transgenic cells.
  • methods comprise delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide
  • the three vectors When the three vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, some cells will take up the third vector, some cells will take up two different vectors, and some cells will take up all three vectors. Only those cells that take up all three vectors are capable of expressing a full-length functional selectable marker protein, thus only those cells are selected as triple-transgenic cells.
  • methods comprise delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a first central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, (c) a third vector comprising (i) a nucleot
  • the four vectors When the four vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, some cells will take up the third vector, some will take up the fourth vector, some cells will take up two different vectors, some cells will take up three different vectors, and some will take up all four vectors. Only those cells that take up all four vectors are capable of expressing a full-length functional selectable marker protein, thus only those cells are selected as quadruple-transgenic cells.
  • FIGS. 1A-1B Split selectable marker for antibiotic co-selection of two separate transgenic vectors.
  • FIG. 1A The coding sequence of a selectable marker is split into an N-terminal fragment (MarN) and a C-terminal fragment (kerC) and separately cloned upstream of an N-terminal fragment of a split intein (IntN) and downstream of a C-terminal fragment of the split intein (IntC), respectively, on two different vectors each carrying a different transgene. These vectors are delivered to cells yielding sub-populations of cells containing either one of the vectors or both of the vectors.
  • FIGS. 2A-2F Details of the split points of Intein- split resistance (Intres) genes (also referred to as selectable marker genes) and plasmids.
  • FIG. 2A Split points for hygromycin resistance protein (SEQ ID NO: 1). Amino acid sequence of hygromycin resistance protein is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. " A C” indicates an insertion of a Cysteine.
  • FIG. 2B Split points for puromycin resistance protein (SEQ ID NO: 2). Amino acid sequence of puromycin resistance protein is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers
  • FIG. 2C Split points for neomycin resistance protein (SEQ ID NO: 3). Amino acid sequence of neomycin resistance gene is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. (FIG.
  • FIG. 3 2-markertron hygromycin (Hygro) intein- split resistance (Intres) genes.
  • Top schematic shows the split points tested for hygromycin resistance gene. The last residue of the N- terminal fragment is indicated on top of the lollipops.
  • Circle lollipops represent split points using NpuDnaE intein while square lollipops represent those using SspDnaB intein.
  • Crossed-out and shaded lollipops indicate split pairs that failed to endow cells with hygromycin resistance.
  • the column plot below shows the percentage of double-transgenic cells (BFP+ mCherry+) in the non- selected (white columns) and the selected cultures (blue columns) analyzed by flow cytometry.
  • FIG. 4. 2-markertron puromycin (Puro) Intres genes. Top schematic shows split points tested for puromycin resistance genes while bottom column plots show percentage of double transgenic cells in the non-selected (white columns) and the selected cultures (brown columns).
  • FIG. 5 2-markertron neomycin (Neo) resistance genes. Top schematic shows split points tested for neomycin resistance genes while bottom column plots show percentage of double transgenic cells in the non-selected (white columns) and the selected cultures (orange columns).
  • FIG. 6 2-markertron blasticidin (Blast) Intres genes. Top schematic shows split points tested for blasticidin resistance gene while bottom column plot shows percentages of double transgnic cells in the non-selected (white columns) and the selected (cyan colums) cultures.
  • FIGS. 7A-7C Gateway-compatible lentiviral destination vectors with 2-markertron Intres markers.
  • FIG. 7A Gateway-compatiable lentiviral destination vector kits for each split Intres marker consists of an N- vector and C- vector.
  • N- vector contains viral LTRs, CAGGS promoter, gateway destination cassette AttL, ccdB gene, chloramphenicol resistance gene that allows LR clonase-mediated recombination of Gateway donor vector carrying transgenes, followed by internal ribosomal entry site (IRES) that allows polycistronic expression of the N-markertron.
  • C- vector contains the C-markertron and allows recombination of another transgene.
  • FIG. 7B Gateway-compatible lentiviral destination vectors with 2-markertron Intres markers.
  • TagBFP2 (as transgene 1) and mCherry (as transgene 2) were cloned into the 2-markertron Intres plasmids by Gateway recombination and delivered to cells by lentiviral transduction, followed by antibiotic selection and flow cytometry analysis.
  • Column plot shows the percentage of
  • Hygro 2-markertron hygromycin
  • Puro Puro, brown column
  • neomycin Neo, orange column
  • NLS- GFP (as transgene 1) that labels nucleus with GFP fluorescence and lifeAct-mScarlet (as transgene 2) that labels F-actin with mScarlet fluorescence were recombined into lentiviral vectors expressing full non-split hygromycin resistance gene or lentiviral vectors with 2-markertron hygromycin Intres genes and used to transduce U20S cells to make dual-label cells.
  • Representative fluorescence microscopic images show GFP, mScarlet and merged channels of cells after hygromycin selection for two weeks.
  • FIGS. 8A-8C Split mScarlet for fluorescence-mediated co-selection of two separate transgenic vectors.
  • FIG. 8A 2-markertron mScarlet proteins. Top schematics shows the split points tested for mScarlet. The last residue of the N-terminal fragment is indicated on top of the lollipops.
  • FIG. 8C The column plot below shows the percentage of mScarlet-positive cells of each of the indicated split points. The column plot above shows the percentage of TagBFP+ EGFP+ cells among the PI cells (white columns) and the mScarlet-positive subset of PI cells (red columns).
  • FIGS. 9A-9D Multi-split selectable markers for co-selection of three or more transgenic vectors.
  • a selectable marker is partitioned into three fragments (Mi, M 2 and M 3 ).
  • the first marker fragment (Mi) is fused upstream of the N-terminal fragment of the first split intein (INI).
  • the second marker fragment (M 2 ) is fused downstream of the C-terminal fragment of the first split intein (Ici) and upstream of the N-terminal fragment of the second split intein (IN2).
  • the third marker fragment (M 3 ) is fused downstream of the C-terminal fragment of the second split intein (Id).
  • the first split intein catalyzes the joining of Mi to M 2 while the second split intein catalyzes the joining of M 2 to M 3 , effectively reconstituting the full selectable marker.
  • FIG. 9B A design of a £-split selectable marker via an "intein chain" mechanism. Similar to the 3-split scenario, the selectable marker is partitioned into k fragments, and are reconstituted through protein trans- splicing mediated by intervening split inteins.
  • FIG. 9C Split points identified from 2-split selectable markers were used in combination to produce 3-split selectable markers.
  • FIG. 9D 3-markertron hygromycin (Hygro) Intres. Top schematics shows the split points tested for hygromycin resistance gene, with residue numbers of the last amino acid of the N-terminal fragments indicated above circle or square lollipops, representing NpaDnaE and SspOna inteins, respectively.
  • FIGS. lOA-lOC Gateway-compatible lentiviral destination vectors with 3-markertron hygromycin Intres genes.
  • FIG. 10A Gateway-compatiable lentiviral destination vector with viral LTRs, CAGGS promoter, gateway destination cassette AttL, ccdB gene, chloramphenicol resistance gene that allows LR clonase-mediated recombination of Gateway donor vector carrying transgenes, followed by internal ribosomal entry site (IRES) that allows polycistronic expression of the each of the three 3-split hygromycin markertrons.
  • IRS internal ribosomal entry site
  • TagBFP2 (as transgene 1) and EGFP (as transgene 2) and mCherry (as transgene 3) were cloned into the 3-split Intres plasmids by Gateway recombination and delivered to cells by lentiviral transduction, followed by antibiotic selection and flow cytometry analysis.
  • FIG. IOC Column plot shows the percentage of BFP+GFP+mCherry+ triple-positive cells in the hygromycin selected (blue columns) versus their corresponding nonselective cultures (white columns).
  • FIG. 11 Four-split Hygro intres.
  • Plasmid 115 expresses a markertron created by fusing amino acid 1-89 of Hygromycin resistance gene [Hygro(l-89)] to NpuDnaE(N) and a leuzine Zipper A motif (LZA).
  • Plasmid 116 expresses a markertron created by fusing, from N- to C-termini, Leucine Zipper B motif (LZB)-NpuDnaGEP(C), Hygro(90-200) and SspDnaB(N).
  • Plasmid 117 expresses a markertron created by fusing, from N- to C-termini, SspDnaB(C), Hygro(201-240), NpuDnaE(N)- LZA. Plasmid 118 expresses a markertron created by fusing LZB-NpuDnaGEP(C) to Hygro(241- 341).
  • FIGS. 12A-12E Intres markers allow enrichment of biallelic targeted cells from
  • Targeting construct pairs containing homology arms for AAVS 1 safe harbor locus were designed to contain full length (FL) non-split or split Intres markers and tested for ability to enrich for biallelic targeted cells via antibiotic selection. (FIG.
  • Plasmids 107 and 108 contains FL Neomycin (Neo) resistance gene driven by endogenous PPP1R12C promoter at the AAVS l locus, FL Hygromycin (Hygro) gene and rtTA Dox-respsonsive transactivator driven by an EFla promoter, as well as FL Blasticidin (Blast) expressed as well as EGFP (plasmid 107) and mScarlet (plasmid 108) from a dox-inducible TetO promoter.
  • Plasmid 106 contains Cas9 and an sgRNA targeting the AAVS locus. 2A: self-cleaving 2A peptides.
  • Plasmids 106, 107 and 108 were co-transfected into HEK293T cells, split, and passaged in dox-containing hygromycin, blasticidin or non- selective media for two weeks, and analyzed by flow cytometry to assay efficiency of biallelic targeting.
  • Plasmids 109 and 110 contain similar structure as Plasmids 107 and 108, but having split Blast Intres in place of the FL Blast.
  • Plasmids 111 and 112 contain an EFla-driven FL Blast and TetO-driven FL Hygro, nitroreductase (NTR), fluorescent protein (EGFP or mCherry) separated by 2A peptides.
  • NTR nitroreductase
  • Plasmids 113 and 114 are similar to Plasmids 111 and 112 but with Hygro Intres in place of FL Hygro.
  • FIG. 12E Flow cytometry analysis of cells transfected with Plasmid 106 (Cas9+AAVS-sgRNA) and the indicated targeting construct pairs, two weeks after culturing in dox-containing non-selective media (Selection: None), blasticidin selection media (Blast) and hygromycin selection media (Hygro).
  • an exemplary method of the present disclosure comprises delivering to a population of cells (a) a vector encoding a first selectable marker protein fragment upstream from an N-terminal intein protein fragment and a first transgene of interest, and (b) another vector encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment and a second (e.g., a different) transgene of interest.
  • Some cells of the population will take up a single vector (carrying only a fragment of the intein, a fragment of the selectable marker protein, and a single transgene), while other cells of the population will take up both vectors (and thus both intein fragments, both selectable marker protein fragments, and both transgenes of interest).
  • the intein protein fragments spontaneously and non-covalently assemble (cooperatively fold) into an intein structure to catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length selectable marker protein, which enables specific selection of those double transgenic cells.
  • the selectable marker protein is an antibiotic resistance protein
  • only double-transgenic cells expressing the full-length (functional) antibiotic resistance protein will survive selection in the present of the particular antibiotic.
  • the selectable marker protein is a fluorescent protein
  • only double-transgenic cells expressing the full-length (functional) fluorescent protein will emit a detectable signal such that only those signal-emitting cells are selected.
  • Another exemplary method of the present disclosure comprises delivering to a population of cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the
  • Some cells of the population will take up a single vector (carrying only a fragment of the intein, a fragment of the selectable marker protein, and a single transgene), while other cells of the population will take up two vectors or all three vectors (and thus all intein fragments, all selectable marker protein fragments, and all transgenes of interest).
  • the intein protein fragments spontaneously and non-covalently assemble (cooperatively fold) into an intein structure to catalyze joining of the N-terminal fragment of the selectable marker protein to the central fragment, and the central fragment to the C-terminal fragment of the selectable marker protein to produce a full-length selectable marker protein, which enables specific selection of those triple-transgenic cells.
  • the selectable marker protein is an antibiotic resistance protein
  • only triple-transgenic cells expressing the full-length (functional) antibiotic resistance protein will survive selection in the present of the particular antibiotic.
  • the selectable marker protein is a fluorescent protein, as another example, only triple- transgenic cells expressing the full-length (functional) fluorescent protein will emit a detectable signal such that only those signal-emitting cells are selected.
  • intein intervening protein
  • protein splicing a unique auto-processing event known as protein splicing in which it excises itself out from a larger precursor polypeptide through the cleavage of two peptide bonds and, in the process, ligates the flanking extein (external protein) sequences through the formation of a new peptide bond.
  • This rearrangement occurs post-translationally (or possibly co-translationally), as intein genes are found embedded in frame within other protein- coding genes.
  • intein-mediated protein splicing is spontaneous; it requires no external factor or energy source, only the folding of the intein domain.
  • the precursor protein contains three segments— an N-extein (N-terminal portion of the protein) followed by the intein followed by a C-extein (C-terminal portion of the protein). Following splicing, the resulting protein contains the N-extein linked to the C-extein.
  • inteins There are two types of inteins: czs-splicing inteins are single polypeptides that are embedded in a host protein, whereas trans-splicing inteins (referred to as split inteins) are separate
  • this acyl unit is transferred to the first residue of the C-extein (which is either serine, threonine, or cysteine) to form a branched intermediate.
  • this branched intermediate is cleaved from the intein by a transamidation reaction involving the C-terminal asparagine residue of the intein.
  • This sets up the final step of the process involving an S-N acyl transfer to create a normal peptide bond between the two exteins (Lockless, SW, Muir, TW PNAS 106(27): 10999-11004 (2009)).
  • intein alleles there are at least 70 different intein alleles, distinguished not only by the type of host gene in which the inteins are embedded, but also the integration point within that host gene (Perler, FB Nucleic Acids Res. 30: 383-384 (2002); Pietrokovski, S Trends Genet. 17: 465-472 (2001)).
  • a small fraction (less than 5%) of the identified intein genes encode split inteins.
  • split inteins are transcribed and translated as two separate polypeptides, the N-intein and C-intein, each fused to one extein.
  • intein fragments spontaneously and non-covalently assemble (cooperatively fold) into the canonical intein structure to carry out protein splicing in trans.
  • split inteins are used to catalyze the joining of two fragments (e.g., an N-terminal fragment and a C-terminal fragment) of a selectable marker protein, such as an antibiotic resistance protein or a fluorescent protein to produce a functional, full-length protein (e.g., FIGS. 1A and IB).
  • a selectable marker protein such as an antibiotic resistance protein or a fluorescent protein
  • a split intein may be a natural split intein or an engineered split intein. Natural split inteins naturally occur in a variety of different organisms. The largest known family of split inteins is found within the DnaE genes of at least 20 cyanobacterial species (Caspi J, et al. Mol. Microbiol. 50: 1569-1577 (2003)). Thus, in some embodiments of the present disclosure, a natural split intein is selected from DnaE inteins.
  • DnaE inteins include Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • a split intein is an engineered split intein.
  • Engineered split inteins may be produced from contiguous inteins (where a contiguous intein is artificially split) or may be modified natural split inteins that, for example, promote efficient protein purification, ligation, modification and cyclization (e.g., NpuGEP and CfaGEP, as described by Stevens, AJ PNAS 114(32): 8538-8543 (2017)).
  • Methods for engineering split inteins are described, for example, by Aranko, AS et al. Protein Eng Des Sel. 27(8): 263-271 (2014), incorporated herein by reference.
  • the engineered split intein is engineered from DnaB inteins (Wu, H, et al. Biochim Biophys Acta 1387(1-2): 422-432 (1998)).
  • the engineered split intein may be a S ⁇ p DnaB S 1 intein.
  • the engineered split intein is engineered from GyrB inteins.
  • the engineered split intein may be a SspGyrB S l l intein.
  • the first intein may be the same as the second intein (e.g., both DnaE inteins). In other embodiments, two different inteins may be used (e.g., a DnaE intein and a DnaB intein). In some embodiments, the first intein is a NpaDnaE intein and the second intein is a NpaDnaE intein.
  • Transgenic (e.g., double and/or triple transgenic) cells of the present disclosure are selected based on their expression of a full-length selectable marker protein.
  • a selectable marker protein generally, confers a trait suitable for artificial selection. Examples of selectable marker proteins include antibiotic resistance proteins and fluorescent proteins.
  • An antibiotic resistance gene is a gene encoding a protein that confers resistance to a particular antibiotic or class of antibiotics.
  • antibiotic resistance genes for use in eukaryotic cells include those encoding proteins that confer resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
  • antibiotic resistance genes for use in prokaryotic cells include those encoding proteins that confer resistance to hygromycin, G418, puromycin, phleomycin Dl, blasticidin, kanamycin, spectinomycin, streptomycin, ampicillin, carbenicillin, bleomycin, erythromycin, polymyxin D, tetracycline and chloramphenicol.
  • Hygromycin B is an antibiotic produced by the bacterium Streptomyces hygroscopicus. It is an aminoglycoside that kills bacteria, fungi and higher eukaryotic cells by inhibiting protein synthesis.
  • Hygromycin phosphotransferase (HPT) encoded by the hpt gene (also referred to as the hph or aphlV gene) originally derived from Escherichia coli, detoxifies the aminocyclitol antibiotic hygromycin B.
  • HPT Hygromycin phosphotransferase
  • the selectable marker gene of the present disclosure is the hpt gene.
  • G418 (GENETICIN ® ) is an aminoglycoside antibiotic similar in structure to gentamicin B l. It is produced by Micromonospora rhodorangea. G418 blocks polypeptide synthesis by inhibiting the elongation step in both prokaryotic and eukaryotic cells. Resistance to G418 is conferred by the neo gene from Tn5 encoding an aminoglycoside 3'-phosphotransferase, APT 3' II. G418 is an analog of neomycin sulfate, and has similar mechanism as neomycin. Thus, in some embodiments, the selectable marker gene of the present disclosure is the neo gene.
  • Puromycin is an aminonucleoside antibiotic, derived from Streptomyces alboniger, that causes premature chain termination during translation taking place in the ribosome. Puromycin is selective for either prokaryotes or eukaryotes. Resistance to puromycin is conferred through expression of the puromycin N-acetyl-transferase (pac) gene.
  • pac puromycin N-acetyl-transferase
  • the selectable marker gene of the present disclosure is the pac gene.
  • Phleomycin Dl is a glycopeptide antibiotic and one of the phleomycins from Streptomyces verticillus belonging to the bleomycin family of antibiotics. It is a broad- spectrum antibiotic that is effective against most bacteria, filamentous fungi, yeast, plant, and animal cells. It causes cell death by intercalating into DNA and induces double strand breaks of the DNA. Resistance to phleomycin Dl is conferred by the product of the Sh ble gene first isolated from Streptoalloteichus hindustanus. Thus, in some embodiments, the selectable marker gene of the present disclosure is the Sh ble gene.
  • Blasticidin S is an antibiotic that is produced by Streptomyces griseochromogenes .
  • Blasticidin prevents the growth of both eukaryotic and prokaryotic cells by inhibiting termination step of translation and peptide bond formation (to lesser extent) by the ribosome. Resistance to blasticidin is conferred by at least three different genes: bis (an acetyltransferase) from
  • the selectable marker gene of the present disclosure is the bis gene, the bsr gene, or the bsd gene.
  • Non-limiting examples of fluorescent proteins that may be used as provided herein include TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ⁇ , mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mScarlet, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby,
  • Full-length selectable marker genes are produced by joining in the same cell two selectable marker gene fragments.
  • one of the fragments is an N-terminal fragment (N-extein), while the other fragment is a C-terminal fragment (C-extein).
  • N-extein N-terminal fragment
  • C-extein C-terminal fragment
  • a first antibiotic resistance protein fragment is an N-terminal antibiotic resistance protein fragment
  • a second antibiotic resistance protein fragment is a C-terminal antibiotic resistance protein fragment
  • a first fluorescent protein fragment is an N-terminal fluorescent protein fragment
  • a second fluorescent protein fragment is a C-terminal fluorescent protein fragment.
  • full-length selectable marker genes are produced by joining in the same cell three or more selectable marker gene fragments.
  • one of the fragments is an N-terminal fragment, one or more (e.g., 1, 2, or 3) of the fragments is a central fragment, and one of the fragments is a C-terminal fragment.
  • An N-terminal fragment may be any protein fragment that includes the free amine group (- NH2) of the full-length protein.
  • a C-terminal fragment may be any protein fragment that includes the free carboxyl group (-COOH).
  • a central fragment may be any protein fragment that is located between the N-terminal fragment and the C-terminal fragment of the full-length protein.
  • amino acids 1-89 of the gene encoding hygromycin (a 341 -amino acid protein) may be referred to as the N-terminal protein fragment, while amino acids 90-341 may be referred to as the C-terminal fragment.
  • amino acids 1-200 of the gene encoding hygromycin may be referred to as the N-terminal protein fragment, while amino acids 201-341 may be referred to as the C-terminal fragment.
  • FIG. 6 shows additional examples where amino acids 1-53, 1-240, or 1-292 are considered the N-terminal protein fragments of full length hygromycin containing amino acids 54-341, 241-341, or 293-341 as the respective C-terminal fragments.
  • amino acids 1-52 of the gene encoding hygromycin (a 341 -amino acid protein) may be referred to as the N-terminal protein fragment
  • amino acids 53-89 may be referred to as the central protein fragment
  • amino acids 90-341 may be referred to as the C-terminal fragment.
  • amino acids 1-89 of the gene encoding hygromycin may be referred to as the N-terminal protein fragment
  • amino acids 90-240 may be referred to as the central fragment
  • amino acids 241-341 may be referred to as the C-terminal fragment.
  • the methods and compositions of the present disclosure are used, in some embodiments, to produce multi-transgenic (e.g., double and/or triple transgenic) cells and/or organisms.
  • the methods use one vector that encodes a first molecule (a first molecule of interest) and another vector that encodes a second molecule (a second molecule of interest).
  • the methods use yet another vector that encodes a third molecules of interest.
  • Additional vectors may encode additional molecules of interest.
  • Molecules of interest may be, for example, polypeptides (e.g., proteins and peptides) or polynucleotides (e.g., nucleic acids, such as DNA or RNA).
  • the first molecule (e.g., located on the first vector) is a protein.
  • the second molecule (e.g., located on the second vector) is a protein.
  • the third molecule (e.g., located on the third vector) is a protein. Examples of proteins of interest include, but are not limited to, enzymes, cytokines, transcription factors, hormones, growth factors, blood factors, antigens and antibodies.
  • the first molecule is a peptide.
  • the second molecule is a peptide.
  • the third molecule is a peptide.
  • the first molecule is a messenger RNA (mRNA).
  • mRNA messenger RNA
  • the second molecule is a mRNA.
  • the third molecule is a mRNA.
  • the mRNA in some embodiments, encodes a vaccine or other antigenic molecule.
  • the first molecule is a non-coding RNA (a RNA that does not encode a protein).
  • the second molecule is a non-coding RNA.
  • the third molecule is a non-coding RNA.
  • non-coding RNA include, but are not limited to, RNA interference molecules, such as microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a vector is any nucleic acid that may be used as a vehicle to carry exogenous (foreign) genetic material into a cell.
  • a vector in some embodiments, is a DNA sequence that includes an insert (e.g., transgene) and a larger sequence that serves as the backbone of the vector.
  • Non-limiting examples of vectors include plasmids, viruses/viral vectors, cosmids, and artificial chromosomes, any of which may be used as provided herein.
  • the vector is a viral vector, such as a viral particle.
  • the vector is an RNA-based vector, such as a self- replicating RNA vector.
  • the first vector is a plasmid
  • the second vector is a plasmid
  • the third vector is a plasmid.
  • a vector includes a promoter operably linked to a nucleic acid encoding a fragment of an intein and a fragment of selectable marker protein.
  • a vector also comprises a promoter operably linked to a nucleic acid, such as a transgene, encoding a molecule of interest.
  • one vector (e.g., a first vector) comprises a nucleotide sequence encoding a first selectable marker protein fragment upstream from a nucleotide sequence encoding an N-terminal intein protein fragment
  • the other vector e.g., a second vector
  • This configuration is equivalent to one vector (e.g., a first vector) comprising a nucleotide sequence encoding an N-terminal intein protein fragment downstream from a nucleotide sequence encoding a first selectable marker protein fragment, and the other vector (e.g., a second vector) comprising a second antibiotic resistance protein fragment downstream from a nucleotide sequence encoding a C-terminal intein protein fragment.
  • upstream and downstream refer to relative positions in a nucleic acid. Each nucleic acid has a 5' end and a 3' end, so named for the carbon position on the deoxyribose (or ribose) ring.
  • upstream is toward the 5' end of the coding strand and downstream is toward the 3' end.
  • a first vector comprises a nucleotide sequence encoding an N- terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein
  • a second vector comprises a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein
  • a third vector comprises a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein.
  • This configuration is equivalent to a (a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a first intein, which is downstream from a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, (b) a second vector comprising a nucleotide sequence encoding an N-terminal fragment of a second intein, which is downstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is downstream from a nucleotide sequence encoding a C-terminal fragment of the first intein, and (c) a third vector comprising a C-terminal fragment of the antibiotic resistance protein, which is downstream from a nucleotide sequence encoding a C-terminal fragment of the second intein.
  • Methods of the present disclosure may be used for the production of transgenic cells and organisms by introducing into host cells the vectors (e.g., first and second vectors) described herein.
  • the cells into which the vectors are introduced may be eukaryotic or prokaryotic.
  • the cells are eukaryotic.
  • Examples of eukaryotic cells for use as provided herein include mammalian cells, plant cells (e.g., crop cells), inset cells (e.g., Drosophila) and fungal cells (e.g., Saccharomyces).
  • Mammalian cells may be, for example, human cells (stem cells or cells from an established cell line), primate cells, equine cells, bovine cells, porcine cells, canine cells, feline cells, or rodent cells (e.g., mouse or rat).
  • mammalian cells for use as provided herein include, but are not limited to, Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) 293 cells, HeLa cells, and NSO cells.
  • the cells are prokaryotic.
  • prokaryotic cells for use as provided herein include bacterial cells.
  • Bacterial cells may be, for example, Escherichia spp. (e.g., Escherichia coli), Streptococcus spp. (e.g., Streptococcus pyogenes, Streptococcus viridans, Streptococcus pneumoniae), Neisseria spp. (e.g., Neisseria gibirrhoea, Neisseria meningitidis), Corynebacterium spp. (e.g., Corynebacterium diphtheriae), Bacillis spp.
  • Escherichia spp. e.g., Escherichia coli
  • Streptococcus spp. e.g., Streptococcus pyogenes, Streptococcus viridans, Streptococcus pneumoniae
  • Clostridium spp. e.g., Clostridium tetani, Clostridium perfringens, Clostridium novyii
  • Mycobacterium spp. e.g., Mycobacterium tuberculosis
  • Shigella spp. e.g., Shigella flexneri, Shigella dysenteriae
  • Salmonella spp. e.g., Salmonella typhi, Salmonella enteritidis
  • Klebsiella spp. e.g., Klebsiella pneumoniae
  • Yersinia spp. e.g., Yersinia pestis
  • Serratia spp. e.g., Serratia marcescens
  • Pseudomonas spp. e.g., Pseudomonas aeruginosa, Pseudomonas mallei
  • Eikenella spp. e.g., Eikenella corrodens
  • Vibrio spp. e.g., Vibrio cholera, Vibrio natriegens
  • Legionella spp. e.g., Legionella micdadei, Legionella bozemani
  • Brucella spp. e.g., Brucella abortus
  • Mycoplasma spp. e.g., Mycoplasma pneumoniae
  • Streptomyces spp. e.g. Streptomyces coelicolor
  • Methods of the present disclosure include delivering vectors to a composition comprising cells and maintaining the composition under conditions that permit introduction of nucleic acid (e.g., first, second, and third vector) into the cells and permit nucleic acid expression in the cells to produce eukaryotic cells.
  • Conditions required for the introduction of nucleic acid (e.g., vectors) into cells are well known. These conditions include, for example, transformation (of prokaryotic cells) conditions, transfection (of eukaryotic cells) conditions, transduction (via virus/viral vector) conditions, and electroporation conditions, any of which may be used as provided herein.
  • methods of the present disclosure include transfecting eukaryotic (e.g. mammalian) cells, while in other embodiments, the methods include transforming prokaryotic (e.g., bacterial) cells.
  • transgenic e.g., multi-transgenic cells, such as double, triple, and/or quarduple transgenic cells depends on the type of selectable marker used.
  • the selectable marker protein is an antibiotic resistance protein
  • the selection step may include exposing the cells to a specific antibiotic and selecting only those cells that survive.
  • the selectable marker protein is a fluorescent protein
  • the selection step may include simply viewing the cells under a microscope and selecting cells that fluoresce, or the selection step may include other fluorescent selection methods, such as fluorescence-activated cell sorting (FACS) sorting.
  • FACS fluorescence-activated cell sorting
  • cells are transduced with viral vectors (e.g., viruses) carrying the nucleic acids as described herein.
  • viral vectors e.g., viruses
  • cells prior to transduction (or other transfection methed), cells are seeded, for example, on well plates (e.g., 12-well plates) at a density of lxlO 4 to lxlO 6 per well.
  • well plates e.g., 12-well plates
  • 100 ⁇ . to 500 ⁇ e.g., 100, 150, 200, 250, 300, 350, 400, 450, or 500 ⁇ ⁇ of each viral vector is added to each well.
  • kits that may be used, for example, to produce and screen for transgenic cells and/or organisms.
  • the kits may include any two or more components as described herein.
  • a kit may comprise (a) a first vector comprising a nucleotide sequence encoding a first selectable marker protein fragment upstream from a nucleotide sequence encoding an N-terminal intein protein fragment; and (b) a second vector comprising a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment, wherein the N-terminal intein protein fragment and the C-terminal intein protein fragment catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length antibiotic resistance protein.
  • kits include any two or more components as described herein.
  • a kit may comprise (a) a first vector comprising a nucleotide sequence encoding an N- terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein, (b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and (c) a third vector comprising a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein, wherein the N-terminal fragment and
  • kits further comprise any one or more of the following components: buffers, salts, cloning enzymes (e.g., LR clonase), competent cells (e.g., competent bacterial cells), transfection reagents, antibiotics, and/or instructions for performing the methods described herein.
  • cloning enzymes e.g., LR clonase
  • competent cells e.g., competent bacterial cells
  • transfection reagents e.g., antibiotics, and/or instructions for performing the methods described herein.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest;
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the antibiotic resistance protein to produce a full-length antibiotic resistance protein.
  • paragraph 1 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
  • DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • split intein is an engineered split intein.
  • non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • first and/or second vector is a plasmid vector or a viral vector.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) an N-terminal fragment of a hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the hygB gene and (ii) a second molecule of interest,
  • N-terminal fragment and C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the hygB gene to a protein fragment encoded by the C-terminal fragment of the hygB gene to produce full-length hygromycin B pho spho transferase .
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-89 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 90-341 of SEQ ID NO: 1 ;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-200 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 201-341 of SEQ ID NO: 1 ;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-53 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 54-341 of SEQ ID NO: 1;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-240 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 241-341 of SEQ ID NO: 1;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-292 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 293-341 of SEQ ID NO: 1.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) a N-terminal fragment of a bsr gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the bsr gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the bsr gene to a protein fragment encoded by the C-terminal fragment of the bsr gene to produce full-length blasticidin-S deaminase.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) a N-terminal fragment of a pac gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the pac gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the pac gene to a protein fragment encoded by the C-terminal fragment of the pac gene to produce full-length puromycin N-acetyl-transferase.
  • the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-63 of SEQ ID NO: 2
  • the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 64-199 of SEQ ID NO: 2;
  • the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-119 of SEQ ID NO: 2
  • the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 120-199 of SEQ ID NO: 2;
  • the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-100 of SEQ ID NO: 2
  • the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 101-199 of SEQ ID NO: 2.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) a N-terminal fragment of a neo gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the neo gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the neo gene to a protein fragment encoded by the C-terminal fragment of the neo gene to produce full-length aminoglycoside 3'- pho spho transferase .
  • the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-133 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 134-267 of SEQ ID NO: 3; or
  • the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-194 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 195-267 of SEQ ID NO: 3.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a method comprising delivering to a composition comprising eukaryotic cells
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein to produce a full-length fluorescent protein. 33.
  • the method of paragraph 51 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
  • the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ⁇ , mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagPvFP-T, mCpple, mRuby,
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • split intein is an engineered split intein.
  • non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a first vector comprising (i) an N-terminal fragment of an egfp gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an egfp gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the egfp gene to a protein fragment encoded by the C-terminal fragment of the egfp gene to produce full-length EGFP protein.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) an N-terminal fragment of an mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an mScarlet gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the mScarlet gene to a protein fragment encoded by the C-terminal fragment of the mScarlet gene to produce full-length mScarlet protein.
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-46 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 47-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-48 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 49-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-51 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 52-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-75 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 76-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-122 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 123-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-140 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 141-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-163 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 164-232 of SEQ ID NO: 6.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a eukaryotic cell comprising
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest;
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the antibiotic resistance protein to produce a full-length antibiotic resistance protein.
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • split intein is an engineered split intein.
  • RNA non- coding ribonucleic acid
  • non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a first vector comprising (i) an N-terminal fragment of a hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the hygB gene and (ii) a second molecule of interest,
  • N-terminal fragment and C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the hygB gene to a protein fragment encoded by the C-terminal fragment of the hygB gene to produce full-length hygromycin B pho spho transferase .
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-89 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 90-341 of SEQ ID NO: 1 ;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-200 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 201-341 of SEQ ID NO: 1 ;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-53 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 54-341 of SEQ ID NO: 1 ;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-240 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 241-341 of SEQ ID NO: 1 ;
  • the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-292 of SEQ ID NO: 1
  • the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 293-341 of SEQ ID NO: 1.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a eukaryotic cell comprising
  • a first vector comprising (i) a N-terminal fragment of a bsr gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the bsr gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the bsr gene to a protein fragment encoded by the C-terminal fragment of the bsr gene to produce full-length blasticidin-S deaminase.
  • the protein fragment encoded by the C-terminal fragment of the bsr gene comprises an amino acid sequence identified by amino acids 103-140 of SEQ ID NO: 4.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a eukaryotic cell comprising
  • a first vector comprising (i) a N-terminal fragment of a pac gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the pac gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the pac gene to a protein fragment encoded by the C-terminal fragment of the pac gene to produce full-length puromycin N-acetyl-transferase.
  • the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-63 of SEQ ID NO: 2
  • the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 64-199 of SEQ ID NO: 2;
  • the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-119 of SEQ ID NO: 2
  • the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 120-199 of SEQ ID NO: 2;
  • the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-100 of SEQ ID NO: 2
  • the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 101-199 of SEQ ID NO: 2.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a eukaryotic cell comprising
  • a first vector comprising (i) a N-terminal fragment of a neo gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the neo gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the neo gene to a protein fragment encoded by the C-terminal fragment of the neo gene to produce full-length aminoglycoside 3'- phosphotransferase.
  • the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-133 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 134-267 of SEQ ID NO: 3; or the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-194 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 195-267 of SEQ ID NO: 3.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a eukaryotic cell comprising
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein to produce a full-length fluorescent protein.
  • the cell of paragraph 85 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
  • the cell of paragraph 86 further comprising selecting the transgenic eukaryotic cells that comprise the full-length fluorescent protein.
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
  • split intein is an engineered split intein.
  • RNA non- coding ribonucleic acid
  • non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a eukaryotic cell comprising
  • a first vector comprising (i) an N-terminal fragment of an egfp gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an egfp gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the egfp gene to a protein fragment encoded by the C-terminal fragment of the egfp gene to produce full-length EGFP protein.
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • a eukaryotic cell comprising
  • a first vector comprising (i) an N-terminal fragment of an mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest;
  • a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an mScarlet gene and (ii) a second molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the mScarlet gene to a protein fragment encoded by the C-terminal fragment of the mScarlet gene to produce full-length mScarlet protein.
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-46 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 47-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-48 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 49-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-51 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 52-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-75 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 76-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-122 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 123-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-140 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 141-232 of SEQ ID NO: 6;
  • the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-163 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 164-232 of SEQ ID NO: 6.
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 16
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
  • the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
  • the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9
  • the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
  • composition comprising the cell of any one of paragraph 85-108.
  • a kit comprising
  • N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the antibiotic resistance protein to produce a full-length antibiotic resistance protein.
  • kits of paragraph 110 wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
  • a kit comprising
  • a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the fluorescent protein, wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein to produce a full-length fluorescent protein.
  • the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ⁇ , mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mRuby2, mRuby2,
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • kit of any one of paragraphs 112- 120 further comprising any one or more of the following components: buffers, salts, cloning enzymes, competent cells, transfection reagents, antibiotics, and/or instructions for performing the methods described herein.
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein, and the
  • the method of paragraph 112 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first, second, and third vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) an N-terminal fragment of a hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a central fragment of the hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
  • a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a C-terminal fragment of the hygB gene and (ii) a nucleotide sequence encoding a third molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of the protein fragment encoded by N-terminal fragment of the hygB gene to a protein fragment encoded by the central fragment of the hygB gene
  • the N-terminal fragment and the C- terminal fragment of the second intein catalyze joining of the protein fragment encoded by the central fragment of the hygB gene to the protein fragment encoded by the C-terminal fragment of the hygB gene, to produce a full-length hygromycin B phosphotransferase.
  • a eukaryotic cell comprising:
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
  • a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the antibiotic resistance protein to the C-terminal fragment of the antibiotic resistance protein, to produce a full-length antibiotic resistance protein.
  • RNA ribonucleic acid
  • miRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • composition comprising the eukaryotic cell of any one of paragraph 142- 154.
  • a kit comprising:
  • a first vector comprising a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein
  • a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and
  • N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein
  • the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the antibiotic resistance protein to the C-terminal fragment of the antibiotic resistance protein, to produce a full-length antibiotic resistance protein.
  • the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
  • kit of any one of paragraphs 156- 164, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
  • kit of any one of paragraphs 156- 164, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
  • RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
  • a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a third molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the fluorescent protein to the central fragment of the fluorescent protein
  • the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein, to produce a full-length fluorescent protein
  • the method of paragraph 169 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first, second, and third vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
  • RNA ribonucleic acid
  • non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • a method comprising delivering to eukaryotic cells
  • a first vector comprising (i) an N-terminal fragment of a mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a central fragment of the mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
  • a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a C-terminal fragment of the mScarlet gene and (ii) a nucleotide sequence encoding a third molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of the protein fragment encoded by N-terminal fragment of the mScarlet gene to a protein fragment encoded by the central fragment of the mScarlet gene
  • the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of the protein fragment encoded by the central fragment of the mScarlet gene to the protein fragment encoded by the C-terminal fragment of the mScarlet gene, to produce a full-length mScarlet protein.
  • a eukaryotic cell comprising: (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
  • a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
  • a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a third molecule of interest,
  • N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the fluorescent protein to the central fragment of the fluorescent protein
  • the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein, to produce a full-length fluorescent protein
  • RNA ribonucleic acid
  • miRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • composition comprising the eukaryotic cell of any one of paragraph 185-200.
  • a kit comprising:
  • a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein
  • a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and
  • N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the fluorescent protein to the central fragment of the fluorescent protein
  • the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein, to produce a full-length fluorescent protein
  • the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald,
  • DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
  • kit of any one of paragraphs 202-210, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
  • kit of any one of paragraphs 202-210, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
  • RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
  • miRNA microRNA
  • siRNA short-interfering RNA
  • shRNA short-hairpin RNA
  • kit of any one of paragraphs 202-214 further comprising any one or more of the following components: buffers, salts, cloning enzymes, competent cells, transfection reagents, antibiotics, and/or instructions for performing the methods described herein.
  • a transgenic selection method comprising delivering to a composition comprising eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding a first selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) upstream from a nucleotide sequence encoding an N-terminal intein protein fragment and (ii) a nucleotide sequence encoding a first molecule, and (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) and (ii) a nucleotide sequence encoding a second molecule, wherein the N- terminal intein protein fragment and the C-terminal intein protein fragment catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length selectable marker protein.
  • a transgenic selection method comprising delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleot
  • a transgenic selection method comprising delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a first central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, (c) a third vector comprising (i) a nucleot
  • RNAs non-coding ribonucleic acids
  • non-coding RNAs are selected from microRNAs (miRNAs), antisense RNAs, short-interfering RNAs (siRNAs), and short-hairpin RNAs (shRNAs).
  • miRNAs microRNAs
  • siRNAs antisense RNAs
  • shRNAs short-hairpin RNAs
  • Selectable markers are often used in genetic engineering to isolate cells with desired genotypes [1].
  • antibiotic resistance genes for use in eukaryotic cells and a limited number of fluorescent proteins whose spectra can be unambiguously differentiated by equipment in ordinary laboratories.
  • researchers often run into the problem of not having enough choices of selectable markers if they are to incorporate multiple transgenes into a cell.
  • selection with multiple antibiotics at the same time is often harsh to cells.
  • Selectable marker recycling may provide a work-around, however, requiring multiple rounds of transgenesis, selection and removal of selection markers [2].
  • each markertron is inserted onto a transgenic vector carrying a specific transgene. Delivery of transgenic vectors containing a set of markertrons yield cells harboring a subset or a complete set of the marketrons. Only cells containing a complete set of markertrons produce a fully reconstituted marker protein via protein splicing and thus passes through selection while cells with partial sets of markertrons are eliminated, achieving co-selection of cells containing all intended transgenes.
  • Intres markers To facilitate adoption of Intres markers, we created Gateway-compatible lentiviral vectors for convenient restriction-ligation-independent LR clonase recombination of transgenes 8 (FIG. 7A). We tested the functionality of these vectors by recombining TagBPF2 and mCherry, respectively to the N- and C-Intres vectors and found robust selection of double transgenic cells (FIG. 7B).
  • One potential utility of Intres vectors is to install different fluorescent markers in cells to label different cellular compartments. To explore such utility, we cloned in NLS-GFP and LifeAct- mScarlet 9, which label nucleus and F-actin, respectively, by Gateway recombination to
  • split fluorescent markers can be used for transgene selection.
  • FIG. 8A NpuDnaE split points for mScarlet fluorescent protein
  • FIG. 8B To test whether split fluorescent markers can be used for transgene selection, we screened for NpuDnaE split points for mScarlet fluorescent protein (FIG. 8A) and identified four split points allowing for >96% enrichment of double transgenic cells and three other split points enabling >60% enrichment of double transgenic cells in mScarlet-gated population, compared to ⁇ 20% double transgenic cells in non-gated population.
  • split points identified for 2-markertron Intres genes we set out to engineer higher degree split markers.
  • We tested combinations of splits points to partition a marker gene into three or more markertrons to allow for co-selection of more than two "unlinked" transgenes with one antibiotics (FIGS. 9A-9B).
  • FIG. 10A Gateway compatible lentiviral vectors with these markers.
  • Three sets of these vectors were each tested by recombining TagBFP (as transgene 1), EGFP (as transgene 2) and mCherry (as transgene 3) into the N-, M-, and C-Intres Gateway destination vectors and used to transduce U20S cells, which were then split and cultured in hygromycin selection or non-selective media (FIG. 10B). Two weeks after selection, cells were analyzed by flow cytometry. All three sets of 3-markertron hygromycin Intres plasmids support triple transgenic cell selection of >99% compared to ⁇ 25% in the non-selected cultures (FIG. IOC).
  • NpuDnaE intein an enhanced variant of NpuDnaE intein known as NpuDnaGEP 10 fused with leucine zipper motifs 11 in combination with the SspDnaB intein. While transduction of all four plasmids containing constituent markertrons produced cells that survived hygromycin selection, leave-one- out transduction did not yield any survival (Table 2).
  • CRISPPv/Cas has recently emerged as a powerful technology for genome engineering and editing.
  • gene knockout based on NHEJ-mediated insertions/deletions occur at high frequency, precise editing and knock-in based on homology directed repair (HDR) using exogenous repair templates (a.k.a targeting constructs) are inefficient.
  • HDR homology directed repair
  • split selectable markers can be incorporated into lentiviral vectors or gene targeting constructs in CRISPR/Cas9 genome editing experiments to enable enrichment of cells with double transgenesis or biallelic knock-ins.
  • 3- and 4-split markers can be generated to allow higher degree transgenic selection. Future development of even higher-degree split selectable markers may enable "hyper-engineering" of cells containing tens of transgenes or targeted knock- ins.
  • the markertron-ORF Gateway donor plasmids were generated either by a nested fusion PCR procedure to combine intein with the coding sequence of fragments of the selectable marker followed by insertion into the pCR8-GW-TOPO plasmid by sequence- and ligation-independent cloning (SLIC) (Li, M.Z. & Elledge, S.J. SLIC: a method for sequence-and ligation-independent cloning.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • Gibco Glutamax
  • Sodium Pyruvate Gibco
  • penicillin-streptomycin Gibco
  • a viral packaging mix of pLPl, pLP2, and VSV-G were co-transfected with each lentiviral vector into Lenti-X 293T cells (ClonTech), seeded the day before in 6-well plates at a concentration of 1.2xl0 6 cells per well, using Lipofectamine 3000. Media was changed 6h after transfection then incubated overnight. 28 hour post transfection, the media supernatant containing virus was filtered using 45uM PES filters then stored at -80°C until use.
  • target cells HEK293T, MCF7, U2-OS
  • media Prior to transduction, media was changed to media containing 10 ⁇ g/mL polybrene, 1 mL per well. 250 ⁇ ⁇ of each respective virus (500 ⁇ ⁇ total for experimental samples with two viruses added) was added to each well and incubated overnight. Media was changed 24 hour post infection. 4 day post infection cells were split into duplicate plates. 5 day post infection media with antibiotic (hygromycin) was added to each respective well of one replicate plate (the other remained under no selection). Antibiotic selection continued for 2 weeks before analysis on FACS.
  • antibiotic hygromycin
  • IKIATRKYLGKQNVYDIGVERDHNFALKNGFIASN (SEQ ID NO: 8)
  • VKVIGRRSLGVQRIFDIGLPQDHNFLLANGAIAAN SEQ ID NO: 17
  • Plasmid 3 pLX-Hygro(l-89)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 4 pLX-NpuDnaE(C)-Hygro(90-341)-IRES-mCherry
  • Plasmid 5 pLX-Hygro(l-200)-SspDnaB(N)-IRES-TagBFP2
  • Plasmid 6 pLX-SspDnaB(C)-Hygro(201-341)-IRES-mCherry
  • Plasmid 7 pLX-Hygro(l-52)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 21 pLX-Neo(l-133)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 22 pLX-NpuDnaE(C)-Neo(134-267)-IRES-mCherry
  • Plasmid 23 pLX-Neo( 1 - 194)-NpuDnaE(N)-IRES -TagBFP2
  • Plasmid 24 pLX-NpuDnaE(C)-Neo( 195-267)-IRES-mCherry
  • Plasmid 27 pCR8-BsaI->ccdbCam ⁇ -BsaI-NpuDnaE(N)-MDl-68-15 (SEQ ID NO:64)
  • Plasmid 28 pCR8-NpuDnaE(C)_BsaI->ccdbCam ⁇ -BsaI-MDl-68-18 (SEQ ID NO:65)
  • Plasmid 29 pCR8-BsaI->ccdbCam ⁇ -BsaI-SspDnaE(N)-MDl-68-12 (SEQ ID NO:66)
  • Plasmid 30 pCR8-SspDnaE(C)_BsaI->ccdbCam ⁇ -BsaI-MDl-68-13 (SEQ ID NO:67)
  • Plasmid 31 pCR8-BsaI->ccdbCam ⁇ -BsaI-SspDnaB(N-S0)-25-135-18 (SEQ ID NO:68)
  • Plasmid 32 pCR8-SspDnaB(C-S0)_BsaI->ccdbCam ⁇ -BsaI-25-
  • Plasmid 33 pLX-mScarlet(l-46)-NpuDnaE(N)_LZA-IRES-TagBFP2
  • Protein mScarlet(l-46)-NpuDnaE(N)_LZA
  • Plasmid 35 pLX-mScarlet(l-48)-NpuDnaE(N)_LZA-IRES-TagBFP2
  • Protein mScarlet(l-48)-NpuDnaE(N)_LZA
  • Plasmid 36 pLX-LZB_NpuDnaE(C)-mScarlet(insCys;49-232)-IRES-GFP
  • Plasmid 37 pLX-mScarlet(l-51)-NpuDnaE(N)_LZA -IRES-TagBFP2
  • Protein mScarlet(l-51)-NpuDnaE(N)_LZA
  • Plasmid 47 pCR8-TagBFP2
  • Plasmid 48 pCR8-mCherry
  • Plasmid 51 pLX-[TagBFP2]-IRES-Hygro(l-89)-NpuDnaE(N) Vector sequence (SEQ ID NO: 106)
  • Plasmid 52 pLX-[mCherry]-IRES-NpuDnaE(C)-Hygro(90-341) Vector sequence (SEQ ID NO: 107)
  • Plasmid 57 pLX-DEST-IRES-Neo(l-194)-NpuDnaE(N)
  • Plasmid 58 pLX-DEST-IRES-NpuDnaE(C)-Neo(195-267)
  • Plasmid 59 pLX-[TagBFP2]-IRES-Neo(l-194)-NpuDnaE(N)
  • Plasmid 60 pLX-[mCherry]-IRES-NpuDnaE(C)-Neo(195-267)
  • Plasmid 61 pLX-mScarlet(l-51)-NpuDnaE(N)-LZA-IRES-TagBFP2
  • Protein mScarlet(l-51)-NpuDnaE(N)-LZA
  • Plasmid 63 pLX-LZB-NpuDnaE(C)-mScarlet( A C; 164-232)-IRES-EGFP
  • Plasmid 64 pLX-Hygro(l-69)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 65 pLX-NpuDnaE(C)-Hygro( A C;70-341)-IRES-mCherry
  • Plasmid 76 pLX-Puro(l-32)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 77 pLX-NpuDnaE(C)-Piiro( A C;33-199)-IRES-mCherry
  • Plasmid 78 pLX-Puro( l -84)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 79 pLX-NpuDnaE(C)-Puro( A C;85-199)-IRES-mCherry
  • Plasmid 80 pLX-Puro(l-137)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 81 pLX-NpuDnaE(C)-Puro( A C; 138-199)-IRES-mCherry
  • Plasmid 82 pLX-Puro(l- 158)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 84 pLX-Puro(l -180)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 86 pLX-Blast( l-58)-NpuDnaE(N)-IRES-TagBFP2
  • Plasmid 91 pLX-Hygro(l-200)-SspDnaB(N-S0)-IRES-TagBFP2
  • Plasmid 94 pLX-DEST-IRES-TagBFP2 (SEQ ID NO: 186)
  • Plasmid 95 pLX-DEST-IRES-EGFP (SEQ ID NO: 187)
  • Plasmid 96 pLX-DEST-IRES-mCherry (SEQ ID NO: 188)
  • Plasmid 97 pLX-Hygro-IRES-TagBFP2
  • Plasmid 98 Plasmid 98: pLX-Hygro-IRES-mCherry
  • Plasmid 99 pLX-Puro-IRES-TagBFP2
  • Plasmid 100 pLX-Puro-IRES-mCherry
  • Plasmid 101 pLX-Hygro-IRES-EGFP
  • Plasmid 102 pLX-NLS_GFP-IRES-Hygro Vector sequence (SEQ ID NO: 194)
  • Plasmid 103 pLX-LifeAct_mCherry-IRES-Hygro
  • Plasmid 104 pLX-NLS_GFP-IRES-Hygro(l-89)-NpuDnaE(N)
  • Plasmid 105 pLX-LifeAct_mScarlet-IRES- NpuDnaE(C)-Hygro(90-341)
  • Plasmid 106 pX330-AAVS l
  • sgRNA spacer sequence gACCCCACAGTGGGGCCACTA (First g does not match genome) (SEQ ID NO: 198)
  • Vector sequence SEQ ID NO: 199
  • Plasmid 107 pAAVS l -Nst-EFl aHygro2ArtTA3(-)_TetO-Blast-P2A-EGFP
  • Plasmid 108 pAAVS l -Nst-EFl aHygro2ArtTA3(-)_TetO-Blast-P2A-mScarlet
  • Plasmid 109 pAAVS l -Nst-EFlaHygro2ArtTA3(-)_TetO-Blast(l-102)_NpuDnaE(N)-P2A-EGFP
  • Plasmid 110 Plasmid 110: pAAVS l -Nst-EFlaHygro2ArtTA3(-)_TetO-NpuDnaE(C)_Blast(103-140)-P2A-mScarlet Vector sequence (SEQ ID NO:203)
  • Plasmid 111 pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO-Hygro-P2A-NTR-E2A-EGFP
  • Plasmid 112 pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO-Hygro-P2A-NTR-E2A-mCherry
  • Plasmid 113 pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO- Hygro(l-89)-NpuDnaE(N)-P2A-NTR-E2A-EGFP Vector sequence (SEQ ID NO:206)
  • Plasmid 114 pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO- NpuDnaE(C)-Hygro(90-341)-P2A-NTR-E2A-mCherry Vector sequence (SEQ ID NO:207)
  • Plasmid 115 pLX-Hygro(l-89)_NpuDnaE(N)_LZA-IRES-TagBFP2
  • Plasmid 116 pLX-LZB_NpuDnaGEP(C)_Hygro(90-200)_SspDnaB(N-S0)-IRES-GFP
  • Plasmid 117 pLX-SspDnaB(C-S0)_Hygro(201-240)_NpuDnaE(N)_LZA-IRES-GFP
  • Plasmid 118 pLX-LZB_NpuDnaGEP(C)_Hygro(241-341)-IRES-mCherry

Abstract

The present disclosure provides a split intein selectable marker system for the production and selection of transgenic cells.

Description

TRANSGENIC SELECTION METHODS AND COMPOSITIONS
RELATED APPLICATIONS
This application claims the benefit under 35 U.S.C. § 119(e) of U.S. provisional application number 62/616,281, filed January 11, 2018, U.S. provisional application number 62/608,478, filed December 20, 2017, U.S. provisional application number 62/624,629, filed January 31, 2018, U.S. provisional application number 62/571,672, filed October 12, 2017, which is incorporated by reference herein in its entirety.
SEQUENCE LISTING
This application contains a Sequence Listing in computer readable form (filename:
J022770007WO00-SEQ-HJD; 1.50 MB - ASCII text file; created October 3, 2018), which incorporated herein by reference in its entirety and forms part of the disclosure. BACKGROUND
Selectable markers are widely adopted in transgenesis and genome editing for selecting engineered cells with a desired genotype. Antibiotic resistance genes (encoding antibiotic resistance proteins) provide resistance to specific antibiotics so that only cells expressing these resistance genes survive and multiply. Antibiotic resistance genes/antibiotics available for use in eukaryotic cells include /zygfi/Hygromycin, neol Geneticin®/G418, /?ac/Puromycin, Sh Wa/Phleomycin Dl (Zeocin), and fed/Blasticidin. Fluorescent proteins, such as green fluorescent protein (GFP) provide another means of cell selection, for example, via fluorescent- activated cell sorting (FACS) techniques or fluorescent microscopy.
SUMMARY
There is a limited number of antibiotic resistance genes/antibiotics available for use in eukaryotic (e.g., mammalian) cells, thus selection schemes for identifying cells containing multiple transgenes are limited. Not only is there a limited number of distinct genes that confer antibiotic resistance in eukaryotic cells, but simultaneous use of as few as three different antibiotic resistance genes can adversely affect the health of transgenic cells. While antibiotic selection can be performed serially, this process is time-consuming. These limitations on selections schemes for identifying transgenic cells are problematic when there is a need to identify cells into which multiple transgenes have been introduced (e.g., to generate a transgenic organism, e.g., animal model, such as a mouse model). Provided herein are methods, compositions and kits useful for the production and/or identification of, for example, cells and/or organisms harboring two or more transgenes (e.g., double-transgenics, triple-transgenics, etc.). For example, the compositions and kits may be used for the production and/or identification of cells and/or organisms harboring two, three, or four transgenes. This technology is based, at least in part, on a protein splicing mechanism initiated by an intein auto-processing domain, which facilitates the joining (conjugation) specifically in multi- transgenic cells of multiple (e.g., two, three, or four) separate selectable marker protein fragments (double-transgenic cells, triple-transgenic eels, or quadruple-transgenic cells). Joining of the two, three, four, or more separate selectable marker protein fragments in the multi-transgenic cells produces a full-length selectable marker protein that confers, for example, antibiotic resistance (an antibiotic resistance protein) or is capable of fluorescence under an appropriate wavelength of light (fluorescent protein). Cells expressing a full-length antibiotic resistance gene survive in the presence of the corresponding antibiotic and thus are selected as multi-transgenic (e.g., double- transgenic, triple-transgenic, or quadruple-transgenic) cells. Likewise, cells expressing a full-length functioning fluorescent protein fluoresce under the appropriate wavelength of light and thus are selected as multi-transgenic (e.g., double-transgenic, triple-transgenic, or quadruple-transgenic) cells.
Thus, the present disclosure provides, in some embodiments, methods comprising delivering to a composition comprising eukaryotic cells two or more vectors, wherein each vector comprises (i) a nucleotide sequence encoding a selectable marker protein fragment linked to an N-terminal intein protein fragment and/or a C-terminal intein protein fragment and (ii) a nucleotide sequence encoding a molecule of interest, wherein the intein protein fragments, when joined in frame to form full-length function proteins, catalyze joining of the selectable marker protein fragments to produce a full-length selectable marker protein. For example, when two vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, and some cells will take up both vectors. Only those cells that take up both vectors are capable of expressing a full-length
functioning selectable marker protien, thus only those cells are selected as double-transgenic cells.
In some embodiments, methods herein comprising delivering to a composition comprising eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding a first selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) upstream from a nucleotide sequence encoding an N-terminal intein protein fragment and (ii) a nucleotide sequence encoding a first molecule, and (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) and (ii) a nucleotide sequence encoding a second molecule, wherein the N- terminal intein protein fragment and the C -terminal intein protein fragment catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length selectable marker protein. When the two vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, and some cells will take up both vectors. Only those cells that take up both vectors are capable of expressing a full-length functioning selectable marker protein, thus only those cells are selected as double-transgenic cells.
In other embodiments, methods comprise delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the central fragment of the selectable markerprotein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the selectable markerprotein to the C-terminal fragment of the selectable markerprotein, to produce a full-length selectable markerprotein. When the three vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, some cells will take up the third vector, some cells will take up two different vectors, and some cells will take up all three vectors. Only those cells that take up all three vectors are capable of expressing a full-length functional selectable marker protein, thus only those cells are selected as triple-transgenic cells.
In still other embodiments, methods comprise delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a first central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a second central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a third intein and (ii) a nucleotide sequence encoding a third molecule of interest, and (d) a fourth vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the third intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the first central fragment of the selectable marker protein, the N-terminal fragment and the C- terminal fragment of the second intein catalyze joining of first central fragment of the selectable marke rprotein to the second central fragment of the selectable marker protein, the N-terminal fragment and the C-terminal fragment of the third intein catalyze joining of second central fragment of the selectable marker protein to the C-terminal fragment of the selectable marker protein to produce a full-length selectable marker protein. When the four vectors are delivered to a population of cells (e.g., under transfection conditions), some cells will take up the first vector (the vector is introduced in the cells), some cells will take up the second vector, some cells will take up the third vector, some will take up the fourth vector, some cells will take up two different vectors, some cells will take up three different vectors, and some will take up all four vectors. Only those cells that take up all four vectors are capable of expressing a full-length functional selectable marker protein, thus only those cells are selected as quadruple-transgenic cells.
It should be understood that any one embodiment described herein, including those only disclosed in the examples or one section of the specification, is intended to be able to combine with any one or more other embodiments unless explicitly disclaimed.
BRIEF DESCRIPTION OF THE DRAWINGS FIGS. 1A-1B. Split selectable marker for antibiotic co-selection of two separate transgenic vectors. (FIG. 1A) The coding sequence of a selectable marker is split into an N-terminal fragment (MarN) and a C-terminal fragment (kerC) and separately cloned upstream of an N-terminal fragment of a split intein (IntN) and downstream of a C-terminal fragment of the split intein (IntC), respectively, on two different vectors each carrying a different transgene. These vectors are delivered to cells yielding sub-populations of cells containing either one of the vectors or both of the vectors. Only in cells with both vectors expressing the two intein- split selectable marker fragments ("markertrons") undergo protein trans- splicing to reconstitute a full-length selectable marker, allowing specific selection and enrichment of the double transgenic cells. (FIG. IB) To screen for split points compatible for inteins for an antibiotic resistance gene, we identified potential split points according to the junctional requirement for the type of intein tested, then cloned the corresponding N-terminal and C-terminal fragments to the split intein scaffolds on lentiviral vectors equipped with TagBFP2 or mCherry fluorescent proteins, which serve as our test transgenes to evaluate selection efficiency. These are delivered into cells via lentiviral transduction. The cells were then split into replicate plates, one subjected to antibiotic selection while the other maintained in non-selective media. Following antibiotic selection, the replicate cultures were analyzed by flow cytometry.
FIGS. 2A-2F. Details of the split points of Intein- split resistance (Intres) genes (also referred to as selectable marker genes) and plasmids. (FIG. 2A) Split points for hygromycin resistance protein (SEQ ID NO: 1). Amino acid sequence of hygromycin resistance protein is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. "AC" indicates an insertion of a Cysteine. (FIG. 2B) Split points for puromycin resistance protein (SEQ ID NO: 2). Amino acid sequence of puromycin resistance protein is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers
corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. "AC" indicates an insertion of a Cysteine. (FIG. 2C) Split points for neomycin resistance protein (SEQ ID NO: 3). Amino acid sequence of neomycin resistance gene is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. (FIG. 2D) Split points for blasticidin resistance protein (SEQ ID NO: 4). Amino acid sequence of blasticidin resistance gene is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. (FIG. 2E) Split points for green fluorescent protein (SEQ ID NO: 5). (FIG. 2F) Split points for mScarlet fluorescent protein (SEQ ID NO: 6). Amino acid sequence of mScarlet gene is presented with clouds labeling the split points characterized in this study. Within the label, the top row indicates the plasmid numbers
corresponding to Table 1. The bottom row indicates the residue number of the last amino acid in the N-terminal fragment, the species of the intein used, and the residue number of the first amino acid in the C-terminal fragment. "AC" indicates an insertion of a Cysteine.
FIG. 3. 2-markertron hygromycin (Hygro) intein- split resistance (Intres) genes. Top schematic shows the split points tested for hygromycin resistance gene. The last residue of the N- terminal fragment is indicated on top of the lollipops. Circle lollipops represent split points using NpuDnaE intein while square lollipops represent those using SspDnaB intein. Crossed-out and shaded lollipops indicate split pairs that failed to endow cells with hygromycin resistance. The column plot below shows the percentage of double-transgenic cells (BFP+ mCherry+) in the non- selected (white columns) and the selected cultures (blue columns) analyzed by flow cytometry.
FIG. 4. 2-markertron puromycin (Puro) Intres genes. Top schematic shows split points tested for puromycin resistance genes while bottom column plots show percentage of double transgenic cells in the non-selected (white columns) and the selected cultures (brown columns).
FIG. 5. 2-markertron neomycin (Neo) resistance genes. Top schematic shows split points tested for neomycin resistance genes while bottom column plots show percentage of double transgenic cells in the non-selected (white columns) and the selected cultures (orange columns).
FIG. 6. 2-markertron blasticidin (Blast) Intres genes. Top schematic shows split points tested for blasticidin resistance gene while bottom column plot shows percentages of double transgnic cells in the non-selected (white columns) and the selected (cyan colums) cultures.
FIGS. 7A-7C. Gateway-compatible lentiviral destination vectors with 2-markertron Intres markers. (FIG. 7A) Gateway-compatiable lentiviral destination vector kits for each split Intres marker consists of an N- vector and C- vector. N- vector contains viral LTRs, CAGGS promoter, gateway destination cassette AttL, ccdB gene, chloramphenicol resistance gene that allows LR clonase-mediated recombination of Gateway donor vector carrying transgenes, followed by internal ribosomal entry site (IRES) that allows polycistronic expression of the N-markertron. Similarly, C- vector contains the C-markertron and allows recombination of another transgene. (FIG. 7B)
TagBFP2 (as transgene 1) and mCherry (as transgene 2) were cloned into the 2-markertron Intres plasmids by Gateway recombination and delivered to cells by lentiviral transduction, followed by antibiotic selection and flow cytometry analysis. Column plot shows the percentage of
BFP+mCherry+ double-positive cell in the selected culture from the 2-markertron hygromycin (Hygro, blue column), puromycin (Puro, brown column), and neomycin (Neo, orange column) experiments versus their corresponding non-selective cultures (white columns). (FIG. 7C) NLS- GFP (as transgene 1) that labels nucleus with GFP fluorescence and lifeAct-mScarlet (as transgene 2) that labels F-actin with mScarlet fluorescence were recombined into lentiviral vectors expressing full non-split hygromycin resistance gene or lentiviral vectors with 2-markertron hygromycin Intres genes and used to transduce U20S cells to make dual-label cells. Representative fluorescence microscopic images show GFP, mScarlet and merged channels of cells after hygromycin selection for two weeks.
FIGS. 8A-8C. Split mScarlet for fluorescence-mediated co-selection of two separate transgenic vectors. (FIG. 8A) 2-markertron mScarlet proteins. Top schematics shows the split points tested for mScarlet. The last residue of the N-terminal fragment is indicated on top of the lollipops. (FIG. 8B) To screen for NpuDnaE intein-compatible split points for mScarlet, we identified potential split points according to the junctional requirement for NpaDnaE intein, then cloned the corresponding N-terminal and C-terminal fragments to the split inteins scaffolds on lentiviral vectors equipped with TagBFP2 or EGFP fluorescent proteins, which serve as our test transgenes to evaluate the selection efficiency. These are delivered into cells via lentiviral transduction. Cells with both lentiviruses contain the necessary protein splicing machinery and mScarlet fragments to reconstitute the full-length mScarlet fluorescent protein, as well as express both TagBFP2 and EGFP transgenes. Cells were subjected to FACS analysis. Boxed schematic shows an example of FACS analysis of the plasmid pair 33+34. PI population was gated for forward scatter and side scatter for live singlet cells. From those, 17.8% of cells are double positive for TagBFP and EGFP transgenes. When the PI cells were further gated for mScarlet-positive (mCherry channel), 99.4% of cells are double positive for TagBFP and EGFP. (FIG. 8C) The column plot below shows the percentage of mScarlet-positive cells of each of the indicated split points. The column plot above shows the percentage of TagBFP+ EGFP+ cells among the PI cells (white columns) and the mScarlet-positive subset of PI cells (red columns).
FIGS. 9A-9D. Multi-split selectable markers for co-selection of three or more transgenic vectors. (FIG. 9A) A selectable marker is partitioned into three fragments (Mi, M2 and M3). The first marker fragment (Mi) is fused upstream of the N-terminal fragment of the first split intein (INI). The second marker fragment (M2) is fused downstream of the C-terminal fragment of the first split intein (Ici) and upstream of the N-terminal fragment of the second split intein (IN2). The third marker fragment (M3) is fused downstream of the C-terminal fragment of the second split intein (Id). The first split intein catalyzes the joining of Mi to M2 while the second split intein catalyzes the joining of M2 to M3, effectively reconstituting the full selectable marker. (FIG. 9B) A design of a £-split selectable marker via an "intein chain" mechanism. Similar to the 3-split scenario, the selectable marker is partitioned into k fragments, and are reconstituted through protein trans- splicing mediated by intervening split inteins. (FIG. 9C) Split points identified from 2-split selectable markers were used in combination to produce 3-split selectable markers. The corresponding fragments were cloned into lentiviral vectors to result in the 3 -split selectable marker structure and a reporter fluorescent transgene per vector. Cells were then transduced with viruses prepared from these vectors, split into selective or non-selective media. After appropriate selection period, the cultures were analyzed by flow cytometry. (FIG. 9D) 3-markertron hygromycin (Hygro) Intres. Top schematics shows the split points tested for hygromycin resistance gene, with residue numbers of the last amino acid of the N-terminal fragments indicated above circle or square lollipops, representing NpaDnaE and SspOna inteins, respectively. Six 3-markertron hygromycin Intreses were tested, each indicated with a numbered line with circle or square indicating the two split points used for each case. Column plot below shows the percentage of triple transgenic (BFP+ GFP+ mCherry+) cells from the non-selective (white columns) and selective (blue columns) cultures for the 3-markertron hygromycin Intres indicated by the numbers below.
FIGS. lOA-lOC. Gateway-compatible lentiviral destination vectors with 3-markertron hygromycin Intres genes. (FIG. 10A) Gateway-compatiable lentiviral destination vector with viral LTRs, CAGGS promoter, gateway destination cassette AttL, ccdB gene, chloramphenicol resistance gene that allows LR clonase-mediated recombination of Gateway donor vector carrying transgenes, followed by internal ribosomal entry site (IRES) that allows polycistronic expression of the each of the three 3-split hygromycin markertrons. (FIG. 10B) TagBFP2 (as transgene 1) and EGFP (as transgene 2) and mCherry (as transgene 3) were cloned into the 3-split Intres plasmids by Gateway recombination and delivered to cells by lentiviral transduction, followed by antibiotic selection and flow cytometry analysis. (FIG. IOC) Column plot shows the percentage of BFP+GFP+mCherry+ triple-positive cells in the hygromycin selected (blue columns) versus their corresponding nonselective cultures (white columns).
FIG. 11. Four-split Hygro intres. (a) Markertrons of 4-split hygro intres expressed by four different plasmids. Plasmid 115 expresses a markertron created by fusing amino acid 1-89 of Hygromycin resistance gene [Hygro(l-89)] to NpuDnaE(N) and a leuzine Zipper A motif (LZA). Plasmid 116 expresses a markertron created by fusing, from N- to C-termini, Leucine Zipper B motif (LZB)-NpuDnaGEP(C), Hygro(90-200) and SspDnaB(N). Plasmid 117 expresses a markertron created by fusing, from N- to C-termini, SspDnaB(C), Hygro(201-240), NpuDnaE(N)- LZA. Plasmid 118 expresses a markertron created by fusing LZB-NpuDnaGEP(C) to Hygro(241- 341).
FIGS. 12A-12E. Intres markers allow enrichment of biallelic targeted cells from
CRISPR/Cas-mediated knock- in experiments. Targeting construct pairs containing homology arms for AAVS 1 safe harbor locus were designed to contain full length (FL) non-split or split Intres markers and tested for ability to enrich for biallelic targeted cells via antibiotic selection. (FIG. 12A) Plasmids 107 and 108 contains FL Neomycin (Neo) resistance gene driven by endogenous PPP1R12C promoter at the AAVS l locus, FL Hygromycin (Hygro) gene and rtTA Dox-respsonsive transactivator driven by an EFla promoter, as well as FL Blasticidin (Blast) expressed as well as EGFP (plasmid 107) and mScarlet (plasmid 108) from a dox-inducible TetO promoter. Plasmid 106 contains Cas9 and an sgRNA targeting the AAVS locus. 2A: self-cleaving 2A peptides. Plasmids 106, 107 and 108 were co-transfected into HEK293T cells, split, and passaged in dox-containing hygromycin, blasticidin or non- selective media for two weeks, and analyzed by flow cytometry to assay efficiency of biallelic targeting. (FIG. 12B) Plasmids 109 and 110 contain similar structure as Plasmids 107 and 108, but having split Blast Intres in place of the FL Blast. (FIG. 12C) Plasmids 111 and 112 contain an EFla-driven FL Blast and TetO-driven FL Hygro, nitroreductase (NTR), fluorescent protein (EGFP or mCherry) separated by 2A peptides. (FIG. 12D) Plasmids 113 and 114 are similar to Plasmids 111 and 112 but with Hygro Intres in place of FL Hygro. (FIG. 12E) Flow cytometry analysis of cells transfected with Plasmid 106 (Cas9+AAVS-sgRNA) and the indicated targeting construct pairs, two weeks after culturing in dox-containing non-selective media (Selection: None), blasticidin selection media (Blast) and hygromycin selection media (Hygro).
DETAILED DESCRIPTION
Provided herein, in some aspects, are methods of producing transgenic (e.g., multi- transgenic , such as double transgenic or triple transgenic) organisms, into which more than one transgene (or other genetic element) is introduced. As shown in FIG. 1A, an exemplary method of the present disclosure comprises delivering to a population of cells (a) a vector encoding a first selectable marker protein fragment upstream from an N-terminal intein protein fragment and a first transgene of interest, and (b) another vector encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment and a second (e.g., a different) transgene of interest. Some cells of the population will take up a single vector (carrying only a fragment of the intein, a fragment of the selectable marker protein, and a single transgene), while other cells of the population will take up both vectors (and thus both intein fragments, both selectable marker protein fragments, and both transgenes of interest). In cells that take up both vectors, following translation, the intein protein fragments spontaneously and non-covalently assemble (cooperatively fold) into an intein structure to catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length selectable marker protein, which enables specific selection of those double transgenic cells. For example, if the selectable marker protein is an antibiotic resistance protein, only double-transgenic cells expressing the full-length (functional) antibiotic resistance protein will survive selection in the present of the particular antibiotic. If the selectable marker protein is a fluorescent protein, as another example, only double-transgenic cells expressing the full-length (functional) fluorescent protein will emit a detectable signal such that only those signal-emitting cells are selected.
Another exemplary method of the present disclosure comprises delivering to a population of cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a third molecule of interest. Some cells of the population will take up a single vector (carrying only a fragment of the intein, a fragment of the selectable marker protein, and a single transgene), while other cells of the population will take up two vectors or all three vectors (and thus all intein fragments, all selectable marker protein fragments, and all transgenes of interest). In cells that take up all three vectors, following translation, the intein protein fragments spontaneously and non-covalently assemble (cooperatively fold) into an intein structure to catalyze joining of the N-terminal fragment of the selectable marker protein to the central fragment, and the central fragment to the C-terminal fragment of the selectable marker protein to produce a full-length selectable marker protein, which enables specific selection of those triple-transgenic cells. For example, if the selectable marker protein is an antibiotic resistance protein, only triple-transgenic cells expressing the full-length (functional) antibiotic resistance protein will survive selection in the present of the particular antibiotic. If the selectable marker protein is a fluorescent protein, as another example, only triple- transgenic cells expressing the full-length (functional) fluorescent protein will emit a detectable signal such that only those signal-emitting cells are selected.
Inteins
An intein (intervening protein) carries out a unique auto-processing event known as protein splicing in which it excises itself out from a larger precursor polypeptide through the cleavage of two peptide bonds and, in the process, ligates the flanking extein (external protein) sequences through the formation of a new peptide bond. This rearrangement occurs post-translationally (or possibly co-translationally), as intein genes are found embedded in frame within other protein- coding genes. Furthermore, intein-mediated protein splicing is spontaneous; it requires no external factor or energy source, only the folding of the intein domain. In nature, the precursor protein contains three segments— an N-extein (N-terminal portion of the protein) followed by the intein followed by a C-extein (C-terminal portion of the protein). Following splicing, the resulting protein contains the N-extein linked to the C-extein.
There are two types of inteins: czs-splicing inteins are single polypeptides that are embedded in a host protein, whereas trans-splicing inteins (referred to as split inteins) are separate
polypeptides that mediate protein splicing after the intein pieces and their protein cargo associate (see, e.g., Paulus, H Annu Rev Biochem 69:447-496 (2000); and Saleh L, Perler FB Chem Rec 6: 183-193 (2006)). Split inteins catalyze a series of chemical rearrangements that require the intein to be properly assembled and folded. The first step in splicing involves an N-S acyl shift in which the N-extein polypeptide is transferred to the side chain of the first residue of the intein. This is then followed by a trans-(thio)esterification reaction in which this acyl unit is transferred to the first residue of the C-extein (which is either serine, threonine, or cysteine) to form a branched intermediate. In the penultimate step of the process, this branched intermediate is cleaved from the intein by a transamidation reaction involving the C-terminal asparagine residue of the intein. This then sets up the final step of the process involving an S-N acyl transfer to create a normal peptide bond between the two exteins (Lockless, SW, Muir, TW PNAS 106(27): 10999-11004 (2009)).
To date, there are at least 70 different intein alleles, distinguished not only by the type of host gene in which the inteins are embedded, but also the integration point within that host gene (Perler, FB Nucleic Acids Res. 30: 383-384 (2002); Pietrokovski, S Trends Genet. 17: 465-472 (2001)). A small fraction (less than 5%) of the identified intein genes encode split inteins. Unlike the more common contiguous inteins, split inteins are transcribed and translated as two separate polypeptides, the N-intein and C-intein, each fused to one extein. Upon translation, the intein fragments spontaneously and non-covalently assemble (cooperatively fold) into the canonical intein structure to carry out protein splicing in trans. The first two split inteins to be characterized, from the cyanobacteria Synechocystis species PCC6803 (Ssp) and Nostoc punctiforme PCC73102 (Npu), are orthologs naturally found inserted in the a subunit of DNA Polymerase III (DnaE). Npu is especially notable due its remarkably fast rate of protein trans-splicing (ti/2 = 50 s at 30 °C). This half-life is significantly shorter than that of Ssp (tm = 80 min at 30 °C) (Shah, NH et al. J. Am. Chem. Soc. 135: 5839 (2013)).
Herein, split inteins are used to catalyze the joining of two fragments (e.g., an N-terminal fragment and a C-terminal fragment) of a selectable marker protein, such as an antibiotic resistance protein or a fluorescent protein to produce a functional, full-length protein (e.g., FIGS. 1A and IB).
A split intein may be a natural split intein or an engineered split intein. Natural split inteins naturally occur in a variety of different organisms. The largest known family of split inteins is found within the DnaE genes of at least 20 cyanobacterial species (Caspi J, et al. Mol. Microbiol. 50: 1569-1577 (2003)). Thus, in some embodiments of the present disclosure, a natural split intein is selected from DnaE inteins. Non-limiting examples of DnaE inteins include Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
In some embodiments, a split intein is an engineered split intein. Engineered split inteins may be produced from contiguous inteins (where a contiguous intein is artificially split) or may be modified natural split inteins that, for example, promote efficient protein purification, ligation, modification and cyclization (e.g., NpuGEP and CfaGEP, as described by Stevens, AJ PNAS 114(32): 8538-8543 (2017)). Methods for engineering split inteins are described, for example, by Aranko, AS et al. Protein Eng Des Sel. 27(8): 263-271 (2014), incorporated herein by reference. In some embodiments, the engineered split intein is engineered from DnaB inteins (Wu, H, et al. Biochim Biophys Acta 1387(1-2): 422-432 (1998)). For example, the engineered split intein may be a S^p DnaB S 1 intein. In some embodiments, the engineered split intein is engineered from GyrB inteins. For example, the engineered split intein may be a SspGyrB S l l intein.
In some embodiments, wherein triple-transgenics are produced, for example, the first intein may be the same as the second intein (e.g., both DnaE inteins). In other embodiments, two different inteins may be used (e.g., a DnaE intein and a DnaB intein). In some embodiments, the first intein is a NpaDnaE intein and the second intein is a NpaDnaE intein.
Selectable Marker Proteins
Transgenic (e.g., double and/or triple transgenic) cells of the present disclosure are selected based on their expression of a full-length selectable marker protein. A selectable marker protein, generally, confers a trait suitable for artificial selection. Examples of selectable marker proteins include antibiotic resistance proteins and fluorescent proteins.
An antibiotic resistance gene is a gene encoding a protein that confers resistance to a particular antibiotic or class of antibiotics. Non-limiting examples of antibiotic resistance genes for use in eukaryotic cells include those encoding proteins that confer resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin. Non-limiting examples of antibiotic resistance genes for use in prokaryotic cells include those encoding proteins that confer resistance to hygromycin, G418, puromycin, phleomycin Dl, blasticidin, kanamycin, spectinomycin, streptomycin, ampicillin, carbenicillin, bleomycin, erythromycin, polymyxin D, tetracycline and chloramphenicol.
Hygromycin B is an antibiotic produced by the bacterium Streptomyces hygroscopicus. It is an aminoglycoside that kills bacteria, fungi and higher eukaryotic cells by inhibiting protein synthesis. Hygromycin phosphotransferase (HPT), encoded by the hpt gene (also referred to as the hph or aphlV gene) originally derived from Escherichia coli, detoxifies the aminocyclitol antibiotic hygromycin B. Thus, in some embodiments, the selectable marker gene of the present disclosure is the hpt gene.
G418 (GENETICIN®) is an aminoglycoside antibiotic similar in structure to gentamicin B l. It is produced by Micromonospora rhodorangea. G418 blocks polypeptide synthesis by inhibiting the elongation step in both prokaryotic and eukaryotic cells. Resistance to G418 is conferred by the neo gene from Tn5 encoding an aminoglycoside 3'-phosphotransferase, APT 3' II. G418 is an analog of neomycin sulfate, and has similar mechanism as neomycin. Thus, in some embodiments, the selectable marker gene of the present disclosure is the neo gene.
Puromycin is an aminonucleoside antibiotic, derived from Streptomyces alboniger, that causes premature chain termination during translation taking place in the ribosome. Puromycin is selective for either prokaryotes or eukaryotes. Resistance to puromycin is conferred through expression of the puromycin N-acetyl-transferase (pac) gene. Thus, in some embodiments, the selectable marker gene of the present disclosure is the pac gene.
Phleomycin Dl (e.g., ZEOCIN®) is a glycopeptide antibiotic and one of the phleomycins from Streptomyces verticillus belonging to the bleomycin family of antibiotics. It is a broad- spectrum antibiotic that is effective against most bacteria, filamentous fungi, yeast, plant, and animal cells. It causes cell death by intercalating into DNA and induces double strand breaks of the DNA. Resistance to phleomycin Dl is conferred by the product of the Sh ble gene first isolated from Streptoalloteichus hindustanus. Thus, in some embodiments, the selectable marker gene of the present disclosure is the Sh ble gene.
Blasticidin S is an antibiotic that is produced by Streptomyces griseochromogenes .
Blasticidin prevents the growth of both eukaryotic and prokaryotic cells by inhibiting termination step of translation and peptide bond formation (to lesser extent) by the ribosome. Resistance to blasticidin is conferred by at least three different genes: bis (an acetyltransferase) from
Streptoverticillum spp.; bsr (a blasticidin-S deaminase) from Bacillus cereus (other bsr genes are known as well); and bsd (another deaminase) from Aspergillus terreus. Thus, in some
embodiments, the selectable marker gene of the present disclosure is the bis gene, the bsr gene, or the bsd gene.
Non-limiting examples of fluorescent proteins that may be used as provided herein include TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mScarlet, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP. Full-length selectable marker genes, in some embodiments, are produced by joining in the same cell two selectable marker gene fragments. In some embodiments, with reference to any full- length protein, one of the fragments is an N-terminal fragment (N-extein), while the other fragment is a C-terminal fragment (C-extein). Thus, in some embodiments, a first antibiotic resistance protein fragment is an N-terminal antibiotic resistance protein fragment, and a second antibiotic resistance protein fragment is a C-terminal antibiotic resistance protein fragment. In other embodiments, a first fluorescent protein fragment is an N-terminal fluorescent protein fragment, and a second fluorescent protein fragment is a C-terminal fluorescent protein fragment.
In other embodiments, full-length selectable marker genes are produced by joining in the same cell three or more selectable marker gene fragments. In some embodiments, with reference to any full-length protein, one of the fragments is an N-terminal fragment, one or more (e.g., 1, 2, or 3) of the fragments is a central fragment, and one of the fragments is a C-terminal fragment.
An N-terminal fragment may be any protein fragment that includes the free amine group (- NH2) of the full-length protein. A C-terminal fragment may be any protein fragment that includes the free carboxyl group (-COOH). A central fragment may be any protein fragment that is located between the N-terminal fragment and the C-terminal fragment of the full-length protein.
For example, amino acids 1-89 of the gene encoding hygromycin (a 341 -amino acid protein) may be referred to as the N-terminal protein fragment, while amino acids 90-341 may be referred to as the C-terminal fragment. Similarly, with reference to FIG. 5, amino acids 1-200 of the gene encoding hygromycin may be referred to as the N-terminal protein fragment, while amino acids 201-341 may be referred to as the C-terminal fragment. FIG. 6 shows additional examples where amino acids 1-53, 1-240, or 1-292 are considered the N-terminal protein fragments of full length hygromycin containing amino acids 54-341, 241-341, or 293-341 as the respective C-terminal fragments.
As another example, amino acids 1-52 of the gene encoding hygromycin (a 341 -amino acid protein) may be referred to as the N-terminal protein fragment, amino acids 53-89 may be referred to as the central protein fragment, and amino acids 90-341 may be referred to as the C-terminal fragment. Similarly, amino acids 1-89 of the gene encoding hygromycin may be referred to as the N-terminal protein fragment, amino acids 90-240 may be referred to as the central fragment, and amino acids 241-341 may be referred to as the C-terminal fragment.
Transgenes and Other Molecules of Interest
The methods and compositions of the present disclosure are used, in some embodiments, to produce multi-transgenic (e.g., double and/or triple transgenic) cells and/or organisms. Thus, in some embodiments, the methods use one vector that encodes a first molecule (a first molecule of interest) and another vector that encodes a second molecule (a second molecule of interest). In some embodiments, the methods use yet another vector that encodes a third molecules of interest.
Additional vectors (e.g., encoding additional central fragments of a selectable marker protein) may encode additional molecules of interest. Molecules of interest may be, for example, polypeptides (e.g., proteins and peptides) or polynucleotides (e.g., nucleic acids, such as DNA or RNA).
In some embodiments, the first molecule (e.g., located on the first vector) is a protein. In some embodiments, the second molecule (e.g., located on the second vector) is a protein. In some embodiments, the third molecule (e.g., located on the third vector) is a protein. Examples of proteins of interest include, but are not limited to, enzymes, cytokines, transcription factors, hormones, growth factors, blood factors, antigens and antibodies.
In some embodiments, the first molecule is a peptide. In some embodiments, the second molecule is a peptide. In some embodiments, the third molecule is a peptide.
In some embodiments, the first molecule is a messenger RNA (mRNA). In some
embodiments, the second molecule is a mRNA. In some embodiments, the third molecule is a mRNA. The mRNA, in some embodiments, encodes a vaccine or other antigenic molecule.
In some embodiments, the first molecule is a non-coding RNA (a RNA that does not encode a protein). In some embodiments, the second molecule is a non-coding RNA. In some
embodiments, the third molecule is a non-coding RNA. Examples of non-coding RNA include, but are not limited to, RNA interference molecules, such as microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
Vectors
Methods of the present disclosure include the use of at least two or at least three different vectors. A vector is any nucleic acid that may be used as a vehicle to carry exogenous (foreign) genetic material into a cell. A vector, in some embodiments, is a DNA sequence that includes an insert (e.g., transgene) and a larger sequence that serves as the backbone of the vector. Non-limiting examples of vectors include plasmids, viruses/viral vectors, cosmids, and artificial chromosomes, any of which may be used as provided herein. In some embodiments, the vector is a viral vector, such as a viral particle. In some embodiments, the vector is an RNA-based vector, such as a self- replicating RNA vector. In some embodiments, the first vector is a plasmid, the second vector is a plasmid, and/or the third vector is a plasmid. A vector, as provided herein, includes a promoter operably linked to a nucleic acid encoding a fragment of an intein and a fragment of selectable marker protein. In some embodiments, a vector also comprises a promoter operably linked to a nucleic acid, such as a transgene, encoding a molecule of interest.
In some embodiments, one vector (e.g., a first vector) comprises a nucleotide sequence encoding a first selectable marker protein fragment upstream from a nucleotide sequence encoding an N-terminal intein protein fragment, while the other vector (e.g., a second vector) comprises a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second antibiotic resistance protein fragment (see, e.g., FIG. 1A). This configuration is equivalent to one vector (e.g., a first vector) comprising a nucleotide sequence encoding an N-terminal intein protein fragment downstream from a nucleotide sequence encoding a first selectable marker protein fragment, and the other vector (e.g., a second vector) comprising a second antibiotic resistance protein fragment downstream from a nucleotide sequence encoding a C-terminal intein protein fragment. The terms "upstream" and "downstream" refer to relative positions in a nucleic acid. Each nucleic acid has a 5' end and a 3' end, so named for the carbon position on the deoxyribose (or ribose) ring. When considering double- stranded DNA, for example, upstream is toward the 5' end of the coding strand and downstream is toward the 3' end.
In some embodiments, (a) a first vector comprises a nucleotide sequence encoding an N- terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein, (b) a second vector comprises a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and (c) a third vector comprises a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein. This configuration is equivalent to a (a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a first intein, which is downstream from a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, (b) a second vector comprising a nucleotide sequence encoding an N-terminal fragment of a second intein, which is downstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is downstream from a nucleotide sequence encoding a C-terminal fragment of the first intein, and (c) a third vector comprising a C-terminal fragment of the antibiotic resistance protein, which is downstream from a nucleotide sequence encoding a C-terminal fragment of the second intein. Cells
Methods of the present disclosure may be used for the production of transgenic cells and organisms by introducing into host cells the vectors (e.g., first and second vectors) described herein. The cells into which the vectors are introduced may be eukaryotic or prokaryotic. In some embodiments, the cells are eukaryotic. Examples of eukaryotic cells for use as provided herein include mammalian cells, plant cells (e.g., crop cells), inset cells (e.g., Drosophila) and fungal cells (e.g., Saccharomyces). Mammalian cells may be, for example, human cells (stem cells or cells from an established cell line), primate cells, equine cells, bovine cells, porcine cells, canine cells, feline cells, or rodent cells (e.g., mouse or rat). Examples of mammalian cells for use as provided herein include, but are not limited to, Chinese hamster ovary (CHO) cells, human embryonic kidney (HEK) 293 cells, HeLa cells, and NSO cells. In some embodiments, the cells are prokaryotic.
Examples of prokaryotic cells for use as provided herein include bacterial cells. Bacterial cells may be, for example, Escherichia spp. (e.g., Escherichia coli), Streptococcus spp. (e.g., Streptococcus pyogenes, Streptococcus viridans, Streptococcus pneumoniae), Neisseria spp. (e.g., Neisseria gibirrhoea, Neisseria meningitidis), Corynebacterium spp. (e.g., Corynebacterium diphtheriae), Bacillis spp. (e.g., Bacillis anthracis, Bacillis subtilis), Lactobacillus spp., Clostridium spp. (e.g., Clostridium tetani, Clostridium perfringens, Clostridium novyii), Mycobacterium spp. (e.g., Mycobacterium tuberculosis), Shigella spp. (e.g., Shigella flexneri, Shigella dysenteriae),
Salmonella spp. (e.g., Salmonella typhi, Salmonella enteritidis), Klebsiella spp. (e.g., Klebsiella pneumoniae), Yersinia spp. (e.g., Yersinia pestis), Serratia spp. (e.g., Serratia marcescens), Pseudomonas spp. (e.g., Pseudomonas aeruginosa, Pseudomonas mallei), Eikenella spp. (e.g., Eikenella corrodens), Haemophilus spp. (e.g., Haemophilus influenza, Haemophilus ducreyi, Haemophilus aegyptius), Vibrio spp. (e.g., Vibrio cholera, Vibrio natriegens), Legionella spp. (e.g., Legionella micdadei, Legionella bozemani), Brucella spp. (e.g., Brucella abortus), Mycoplasma spp. (e.g., Mycoplasma pneumoniae) or Streptomyces spp. (e.g. Streptomyces coelicolor,
Streptomyces lividans, Streptomyces albus).
Delivery and Selection Methods
Methods of the present disclosure, in some embodiments, include delivering vectors to a composition comprising cells and maintaining the composition under conditions that permit introduction of nucleic acid (e.g., first, second, and third vector) into the cells and permit nucleic acid expression in the cells to produce eukaryotic cells. Conditions required for the introduction of nucleic acid (e.g., vectors) into cells are well known. These conditions include, for example, transformation (of prokaryotic cells) conditions, transfection (of eukaryotic cells) conditions, transduction (via virus/viral vector) conditions, and electroporation conditions, any of which may be used as provided herein. Thus, in some embodiments, methods of the present disclosure include transfecting eukaryotic (e.g. mammalian) cells, while in other embodiments, the methods include transforming prokaryotic (e.g., bacterial) cells.
The selection of transgenic, e.g., multi-transgenic cells, such as double, triple, and/or quarduple transgenic cells depends on the type of selectable marker used. For example, if the selectable marker protein is an antibiotic resistance protein, the selection step may include exposing the cells to a specific antibiotic and selecting only those cells that survive. If the selectable marker protein is a fluorescent protein, the selection step may include simply viewing the cells under a microscope and selecting cells that fluoresce, or the selection step may include other fluorescent selection methods, such as fluorescence-activated cell sorting (FACS) sorting.
In some embodiments, cells are transduced with viral vectors (e.g., viruses) carrying the nucleic acids as described herein. In some embodiments, prior to transduction (or other transfection methed), cells are seeded, for example, on well plates (e.g., 12-well plates) at a density of lxlO4 to lxlO6 per well. In some embodiments 100 μΐ. to 500 μΐ,, e.g., 100, 150, 200, 250, 300, 350, 400, 450, or 500 μΐ^ of each viral vector is added to each well.
Kits
The present disclosure also provides kits that may be used, for example, to produce and screen for transgenic cells and/or organisms. The kits may include any two or more components as described herein. For example, a kit may comprise (a) a first vector comprising a nucleotide sequence encoding a first selectable marker protein fragment upstream from a nucleotide sequence encoding an N-terminal intein protein fragment; and (b) a second vector comprising a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment, wherein the N-terminal intein protein fragment and the C-terminal intein protein fragment catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length antibiotic resistance protein.
In some emnbodiments, the kits include any two or more components as described herein. For example, a kit may comprise (a) a first vector comprising a nucleotide sequence encoding an N- terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein, (b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and (c) a third vector comprising a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the antibiotic resistance protein to the C-terminal fragment of the antibiotic resistance protein, to produce a full-length antibiotic resistance protein.
In some embodiments, the kits further comprise any one or more of the following components: buffers, salts, cloning enzymes (e.g., LR clonase), competent cells (e.g., competent bacterial cells), transfection reagents, antibiotics, and/or instructions for performing the methods described herein. Additional Embodiments
Additional embodiments of the present disclosure are encompassed by the following numbered paragraphs:
1. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest; and
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the antibiotic resistance protein to produce a full-length antibiotic resistance protein.
2. The method of paragraph 1 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
3. The method of paragraph 2 further comprising selecting the transgenic eukaryotic cells that comprise the full-length antibiotic resistance protein.
4. The method of any one of paragraphs 1-3, wherein the eukaryotic cells are mammalian cells. 5. The method of any one of paragraphs 1-4, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
6. The method of any one of paragraphs 1-5, wherein the intein is a split intein.
7. The method of paragraph 6, wherein the split intein is a natural split intein.
8. The method of paragraph 7, wherein the natural split intein is selected from DnaE inteins. 9. The method of paragraph 8, wherein the DnaE inteins are selected from Synechocystis sp.
DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
10. The method of paragraph 6, wherein the split intein is an engineered split intein.
11. The method of paragraph 10, wherein the engineered split intein is engineered from DnaB inteins.
12. The method of paragraph 11, wherein the engineered split intein is a SspOna S 1 intein. 13. The method of paragraph 12, wherein the engineered split intein is engineered from GyrB inteins.
14. The method of paragraph 13, wherein the engineered split intein is a SspGyrB S 11 intein.
15. The method of any one of paragraphs 1-14, wherein the first and/or second molecule is a protein.
16. The method of any one of paragraphs 1-15, wherein the first and/or second molecule is a non-coding ribonucleic acid (RNA).
17. The method of paragraph 16, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
18. The method of any one of paragraphs 1-17, wherein the first and/or second vector is a plasmid vector or a viral vector.
19. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) an N-terminal fragment of a hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the hygB gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the hygB gene to a protein fragment encoded by the C-terminal fragment of the hygB gene to produce full-length hygromycin B pho spho transferase .
20. The method of paragraph 19, wherein the first amino acid of the protein fragment encoded by the second hygB gene fragment is cysteine.
21. The method of paragraph 23, wherein
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-89 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 90-341 of SEQ ID NO: 1 ;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-200 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 201-341 of SEQ ID NO: 1 ;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-53 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 54-341 of SEQ ID NO: 1;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-240 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 241-341 of SEQ ID NO: 1;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-292 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 293-341 of SEQ ID NO: 1.
22. The method of any one of paragraphs 23-21, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
23. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a N-terminal fragment of a bsr gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the bsr gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the bsr gene to a protein fragment encoded by the C-terminal fragment of the bsr gene to produce full-length blasticidin-S deaminase.
24. The method of paragraph 23, wherein the protein fragment encoded by the N-terminal fragment of the bsr gene comprises an amino acid sequence identified by amino acids 1-102 of SEQ ID NO: 4, and the protein fragment encoded by the C-terminal fragment of the bsr gene comprises an amino acid sequence identified by amino acids 103-140 of SEQ ID NO: 4.
25. The method of paragraph 22 or 23, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
26. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a N-terminal fragment of a pac gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the pac gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the pac gene to a protein fragment encoded by the C-terminal fragment of the pac gene to produce full-length puromycin N-acetyl-transferase.
27. The method of paragraph 26, wherein
the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-63 of SEQ ID NO: 2, and the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 64-199 of SEQ ID NO: 2;
the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-119 of SEQ ID NO: 2, and the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 120-199 of SEQ ID NO: 2;
the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-100 of SEQ ID NO: 2, and the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 101-199 of SEQ ID NO: 2.
28. The method of paragraph 26 or 27, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
29. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a N-terminal fragment of a neo gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the neo gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the neo gene to a protein fragment encoded by the C-terminal fragment of the neo gene to produce full-length aminoglycoside 3'- pho spho transferase .
30. The method of paragraph 29, wherein
the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-133 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 134-267 of SEQ ID NO: 3; or
the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-194 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 195-267 of SEQ ID NO: 3.
31. The method of paragraph 29 or 30, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
32. A method comprising delivering to a composition comprising eukaryotic cells
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein to produce a full-length fluorescent protein. 33. The method of paragraph 51 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
34. The method of paragraph 33 further comprising selecting the transgenic eukaryotic cells that comprise the full-length fluorescent protein.
35. The method of any one of paragraphs 32-34, wherein the eukaryotic cells are mammalian cells.
36. The method of any one of paragraphs 32-35, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagPvFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP.
37. The method of any one of paragraphs 32-36, wherein the intein is a split intein.
38. The method of paragraph 37, wherein the split intein is a natural split intein.
39. The method of paragraph 38, wherein the natural split intein is selected from DnaE inteins.
40. The method of paragraph 39, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
41. The method of paragraph 40, wherein the split intein is an engineered split intein.
42. The method of paragraph 41, wherein the engineered split intein is engineered from DnaB inteins.
43. The method of paragraph 42, wherein the engineered split intein is a SspOna S I intein. 44. The method of paragraph 42, wherein the engineered split intein is engineered from GyrB inteins.
45. The method of paragraph 44, wherein the engineered split intein is a SspGyrB S 11 intein.
46. The method of any one of paragraphs 32-45, wherein the first and/or second molecule is a protein.
47. The method of any one of paragraphs 32-46, wherein the first and/or second molecule is a non-coding ribonucleic acid (RNA).
48. The method of paragraph 47, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
49. The method of any one of paragraphs 32-48, wherein the first and/or second vector is a plasmid vector or a viral vector. 50. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) an N-terminal fragment of an egfp gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an egfp gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the egfp gene to a protein fragment encoded by the C-terminal fragment of the egfp gene to produce full-length EGFP protein.
51. The method of paragraph 50, wherein the protein fragment encoded by the N-terminal fragment of the egfp gene comprises an amino acid sequence identified by amino acids 1-175 of SEQ ID NO: 5, and the protein fragment encoded by the C-terminal fragment of the egfp gene comprises an amino acid sequence identified by amino acids 175-239 of SEQ ID NO: 5.
52. The method of paragraph 50 or 51, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
53. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) an N-terminal fragment of an mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an mScarlet gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the mScarlet gene to a protein fragment encoded by the C-terminal fragment of the mScarlet gene to produce full-length mScarlet protein.
54. The method of paragraph 53, wherein
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-46 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 47-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-48 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 49-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-51 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 52-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-75 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 76-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-122 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 123-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-140 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 141-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-163 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 164-232 of SEQ ID NO: 6.
55. The method of paragraph 53 or 54, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
56. A eukaryotic cell, comprising
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest; and
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the antibiotic resistance protein to produce a full-length antibiotic resistance protein.
57. The cell of paragraph 56, wherein the eukaryotic cells are mammalian cells.
58. The cell of paragraph 56 or 57, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
59. The cell of any one of paragraphs 56-58, wherein the intein is a split intein.
60. The cell of paragraph 59, wherein the split intein is a natural split intein.
61. The cell of paragraph 60, wherein the natural split intein is selected from DnaE inteins.
62. The cell of paragraph 61, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
63. The cell of paragraph 59, wherein the split intein is an engineered split intein.
64. The cell of paragraph 63, wherein the engineered split intein is engineered from DnaB inteins.
65. The cell of paragraph 64, wherein the engineered split intein is a SspOna S I intein.
66. The cell of paragraph 65, wherein the engineered split intein is engineered from GyrB inteins.
67. The cell of paragraph 66, wherein the engineered split intein is a SspGyrB S 11 intein.
68. The cell of any one of paragraphs 56-67, wherein the first and/or second molecule is a protein.
69. The cell of any one of paragraphs 56-68, wherein the first and/or second molecule is a non- coding ribonucleic acid (RNA).
70. The cell of paragraph 69, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
71. The cell of any one of paragraphs 56-70, wherein the first and/or second vector is a plasmid vector or a viral vector.
72. A cell comprising
(a) a first vector comprising (i) an N-terminal fragment of a hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the hygB gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the hygB gene to a protein fragment encoded by the C-terminal fragment of the hygB gene to produce full-length hygromycin B pho spho transferase .
73. The cell of paragraph 72, wherein the first amino acid of the protein fragment encoded by the second hygB gene fragment is cysteine.
74. The cell of paragraph 73, wherein
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-89 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 90-341 of SEQ ID NO: 1 ;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-200 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 201-341 of SEQ ID NO: 1 ;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-53 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 54-341 of SEQ ID NO: 1 ;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-240 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 241-341 of SEQ ID NO: 1 ;
the protein fragment encoded by the N-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 1-292 of SEQ ID NO: 1, and the protein fragment encoded by the C-terminal fragment of the hygB gene comprises an amino acid sequence identified by amino acids 293-341 of SEQ ID NO: 1.
75. The cell of any one of paragraphs 72-74, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
76. A eukaryotic cell, comprising
(a) a first vector comprising (i) a N-terminal fragment of a bsr gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the bsr gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the bsr gene to a protein fragment encoded by the C-terminal fragment of the bsr gene to produce full-length blasticidin-S deaminase.
77. The cell of paragraph 76, wherein the protein fragment encoded by the N-terminal fragment of the bsr gene comprises an amino acid sequence identified by amino acids 1-102 of SEQ ID NO:
4, and the protein fragment encoded by the C-terminal fragment of the bsr gene comprises an amino acid sequence identified by amino acids 103-140 of SEQ ID NO: 4.
78. The cell of paragraph 76 or 77, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
79. A eukaryotic cell, comprising
(a) a first vector comprising (i) a N-terminal fragment of a pac gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the pac gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the pac gene to a protein fragment encoded by the C-terminal fragment of the pac gene to produce full-length puromycin N-acetyl-transferase. 80. The cell of paragraph 79, wherein the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-63 of SEQ ID NO: 2, and the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 64-199 of SEQ ID NO: 2;
the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-119 of SEQ ID NO: 2, and the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 120-199 of SEQ ID NO: 2;
the protein fragment encoded by the N-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 1-100 of SEQ ID NO: 2, and the protein fragment encoded by the C-terminal fragment of the pac gene comprises an amino acid sequence identified by amino acids 101-199 of SEQ ID NO: 2.
81. The cell of paragraph 79 or 80, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
82. A eukaryotic cell, comprising
(a) a first vector comprising (i) a N-terminal fragment of a neo gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the neo gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the neo gene to a protein fragment encoded by the C-terminal fragment of the neo gene to produce full-length aminoglycoside 3'- phosphotransferase.
83. The cell of paragraph 82, wherein
the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-133 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 134-267 of SEQ ID NO: 3; or the protein fragment encoded by the N-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 1-194 of SEQ ID NO: 3 and the protein fragment encoded by the C-terminal fragment of the neo gene comprises an amino acid sequence identified by amino acids 195-267 of SEQ ID NO: 3.
84. The cell of paragraph 82 or 83, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
85. A eukaryotic cell, comprising
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein to produce a full-length fluorescent protein.
86. The cell of paragraph 85 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
87. The cell of paragraph 86 further comprising selecting the transgenic eukaryotic cells that comprise the full-length fluorescent protein.
88. The cell of any one of paragraphs 85-87, wherein the eukaryotic cells are mammalian cells.
89. The cell of any one of paragraphs 85-88, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagPvFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP. 90. The cell of any one of paragraphs 85-89, wherein the intein is a split intein.
91. The cell of paragraph 90, wherein the split intein is a natural split intein.
92. The cell of paragraph 91, wherein the natural split intein is selected from DnaE inteins.
93. The cell of paragraph 92, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
94. The cell of paragraph 93, wherein the split intein is an engineered split intein.
95. The cell of paragraph 94, wherein the engineered split intein is engineered from DnaB inteins.
96. The cell of paragraph 95, wherein the engineered split intein is a SspOna S I intein.
97. The cell of paragraph 95, wherein the engineered split intein is engineered from GyrB inteins.
98. The cell of paragraph 97, wherein the engineered split intein is a SspGyrB S 11 intein.
99. The cell of any one of paragraphs 85-98, wherein the first and/or second molecule is a protein.
100. The cell of any one of paragraphs 85-99, wherein the first and/or second molecule is a non- coding ribonucleic acid (RNA).
101. The cell of paragraph 100, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
102. The cell of any one of paragraphs 85- 101, wherein the first and/or second vector is a plasmid vector or a viral vector.
103. A eukaryotic cell, comprising
(a) a first vector comprising (i) an N-terminal fragment of an egfp gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an egfp gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the egfp gene to a protein fragment encoded by the C-terminal fragment of the egfp gene to produce full-length EGFP protein.
104. The cell of paragraph 103, wherein the protein fragment encoded by the N-terminal fragment of the egfp gene comprises an amino acid sequence identified by amino acids 1- 175 of SEQ ID NO: 5, and the protein fragment encoded by the C-terminal fragment of the egfp gene comprises an amino acid sequence identified by amino acids 175-239 of SEQ ID NO: 5.
105. The cell of paragraph 103 or 104, wherein the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
106. A eukaryotic cell, comprising
(a) a first vector comprising (i) an N-terminal fragment of an mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein and (ii) a first molecule of interest; and
(b) a second vector comprising (ii) a nucleotide sequence encoding a C-terminal fragment of an intein, which is upstream from a C-terminal fragment of an mScarlet gene and (ii) a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of a protein fragment encoded by the N-terminal fragment of the mScarlet gene to a protein fragment encoded by the C-terminal fragment of the mScarlet gene to produce full-length mScarlet protein.
107. The cell of paragraph 106, wherein
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-46 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 47-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-48 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 49-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-51 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 52-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-75 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 76-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-122 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 123-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-140 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 141-232 of SEQ ID NO: 6;
the protein fragment encoded by the N-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 1-163 of SEQ ID NO: 6, and the protein fragment encoded by the C-terminal fragment of the mScarlet gene comprises an amino acid sequence identified by amino acids 164-232 of SEQ ID NO: 6.
108. The cell of paragraph 106 or 107, wherein
the N-terminal fragment of the intein is identified by SEQ ID NO: 16, and the C-terminal fragment of the intein is identified by SEQ ID NO: 17;
the N-terminal fragment of the intein is identified by SEQ ID NO:7, and the C-terminal fragment of the intein is identified by SEQ ID NO:8; or
the N-terminal fragment of the intein is identified by SEQ ID NO: 18 or SEQ ID NO:9, and the C-terminal fragment of the intein is identified by SEQ ID NO: 19 or SEQ ID NO: 10.
109. A composition comprising the cell of any one of paragraph 85-108.
110. A kit, comprising
(a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein; and
(b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the antibiotic resistance protein,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the antibiotic resistance protein to produce a full-length antibiotic resistance protein.
111. The kit of paragraph 110, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
112. A kit, comprising
(a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein; and
(b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the fluorescent protein, wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein to produce a full-length fluorescent protein.
113. The kit of paragraph 112, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP.
114. The kit of any one of paragraphs 110- 113, wherein the intein is a split intein.
115. The kit of paragraph 114, wherein the split intein is a natural split intein or an engineered split intein.
116. The kit of paragraph 115, wherein the natural split intein is selected from DnaE inteins.
117. The kit of paragraph 116, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
118. The kit of paragraph 115, wherein the engineered split intein is engineered from DnaB inteins or GyrB inteins.
119. The kit of paragraph 118, wherein the engineered split intein is a SspOna S 1 intein.
120. The kit of paragraph 118, wherein the engineered split intein is a SspGyrB S 11 intein.
121. The kit of any one of paragraphs 112- 120, further comprising any one or more of the following components: buffers, salts, cloning enzymes, competent cells, transfection reagents, antibiotics, and/or instructions for performing the methods described herein.
122. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the antibiotic resistance protein to the C-terminal fragment of the antibiotic resistance protein, to produce a full-length antibiotic resistance protein.
123. The method of paragraph 112 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first, second, and third vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
124. The method of paragraph 123 further comprising selecting the transgenic eukaryotic cells that comprise the full-length antibiotic resistance protein.
125. The method of any one of paragraphs 112-124, wherein the eukaryotic cells are mammalian cells.
126. The method of any one of paragraphs 112-125, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
127. The method of paragraph 126, wherein the antibiotic resistance protein confers resistance to hygromycin.
128. The method of any one of paragraphs 112-127, wherein the first intein is a split intein.
129. The method of any one of paragraphs 112-128, wherein the second intein is a split intein.
130. The method of paragraph 128 or 129, wherein the split intein is a natural split intein.
131. The method of paragraph 130, wherein the natural split intein is selected from DnaE inteins.
132. The method of paragraph 131, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
133. The method of paragraph 132, wherein the first intein is an NpaDnaE intein and the second intein is an NpaDnaE intein.
134. The method of any one of paragraphs 112-133, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
135. The method of any one of paragraphs 112-133, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
136. The method of paragraph 135, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
137. The method of any one of paragraphs 112-136, wherein the first vector, second vector, third vector, or any combination thereof, is a plasmid vector or a viral vector.
138. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) an N-terminal fragment of a hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a central fragment of the hygB gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a C-terminal fragment of the hygB gene and (ii) a nucleotide sequence encoding a third molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of the protein fragment encoded by N-terminal fragment of the hygB gene to a protein fragment encoded by the central fragment of the hygB gene, and the N-terminal fragment and the C- terminal fragment of the second intein catalyze joining of the protein fragment encoded by the central fragment of the hygB gene to the protein fragment encoded by the C-terminal fragment of the hygB gene, to produce a full-length hygromycin B phosphotransferase.
139. The method of paragraph 138, wherein the first vector encodes the sequence identified by SEQ ID NO: 29, the second vector encodes the sequence identified by SEQ ID NO: 61, and the third vector encodes the sequence identified by SEQ ID NO: 23.
140. The method of paragraph 138, wherein the first vector encodes the sequence identified by SEQ ID NO: 21, the second vector encodes the sequence identified by SEQ ID NO: 61, and the third vector encodes the sequence identified by SEQ ID NO: 35.
141. A eukaryotic cell comprising:
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the antibiotic resistance protein to the C-terminal fragment of the antibiotic resistance protein, to produce a full-length antibiotic resistance protein.
142. The eukaryotic cell of paragraph 112, wherein the eukaryotic cells are mammalian cells.
143. The eukaryotic cell of paragraph 141 or 142, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
144. The eukaryotic cell of paragraph 143, wherein the antibiotic resistance protein confers resistance to hygromycin.
145. The eukaryotic cell of any one of paragraphs 141- 144, wherein the first intein is a split intein.
146. The eukaryotic cell of any one of paragraphs 142- 145, wherein the second intein is a split intein.
147. The eukaryotic cell of paragraph 145 or 146 , wherein the split intein is a natural split intein.
148. The eukaryotic cell of paragraph 147, wherein the natural split intein is selected from DnaE inteins.
149. The eukaryotic cell of paragraph 148, wherein the DnaE inteins are selected from
Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
150. The eukaryotic cell of paragraph 149, wherein the first intein is an NpaDnaE intein and the second intein is an NpaDnaE intein.
151. The eukaryotic cell of any one of paragraphs 142- 150, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
152. The eukaryotic cell of any one of paragraphs 142- 150, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
153. The eukaryotic cell of paragraph 152, wherein the non-coding RNA is a microRNA
(miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
154. The eukaryotic cell of any one of paragraphs 142- 153, wherein the first vector, second vector, third vector, or any combination thereof, is a plasmid vector or a viral vector.
155. A composition comprising the eukaryotic cell of any one of paragraph 142- 154.
156. A kit comprising:
(a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of an antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein,
(b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the antibiotic resistance protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and
(c) a third vector comprising a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the antibiotic resistance protein,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the antibiotic resistance protein to the central fragment of the antibiotic resistance protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the antibiotic resistance protein to the C-terminal fragment of the antibiotic resistance protein, to produce a full-length antibiotic resistance protein. 157. The kit of paragraph 156, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
158. The kit of paragraph 157, wherein the antibiotic resistance protein confers resistance to hygromycin.
159. The kit of any one of paragraphs 156- 158, wherein the first intein is a split intein.
160. The kit of any one of paragraphs 156- 159, wherein the second intein is a split intein.
161. The kit of paragraph 159 or 160, wherein the split intein is a natural split intein.
162. The kit of paragraph 161, wherein the natural split intein is selected from DnaE inteins.
163. The kit of paragraph 162, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
164. The kit of paragraph 163, wherein the first intein is an NpaDnaE intein and the second intein is an NpaDnaE intein.
165. The kit of any one of paragraphs 156- 164, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
166. The kit of any one of paragraphs 156- 164, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
167. The kit of paragraph 166, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
168. The kit of any one of paragraphs 156- 167, wherein the first vector, second vector, third vector, or any combination thereof, is a plasmid vector or a viral vector. 169. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a third molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the fluorescent protein to the central fragment of the fluorescent protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein, to produce a full-length fluorescent protein.
170. The method of paragraph 169 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first, second, and third vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
171. The method of paragraph 170 further comprising selecting the transgenic eukaryotic cells that comprise the full-length fluorescent protein.
172. The method of any one of paragraphs 169-171, wherein the eukaryotic cells are mammalian cells.
173. The method of any one of paragraphs 169-172, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP,
Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mScarlet, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP.
174. The method of paragraph 173, wherein the fluorescent protein is mScarlet.
175. The method of any one of paragraphs 169-174, wherein the first intein is a split intein.
176. The method of any one of paragraphs 169-175, wherein the second intein is a split intein. 177. The method of paragraph 175 or 176, wherein the split intein is a natural split intein. 178. The method of paragraph 177, wherein the natural split intein is selected from DnaE inteins.
179. The method of paragraph 178, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
180. The method of paragraph 179, wherein the first intein is an NpaDnaE intein and the second intein is an NpaDnaE intein.
181. The method of any one of paragraphs 169-170, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
182. The method of any one of paragraphs 169-180, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
183. The method of paragraph 182, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
184. The method of any one of paragraphs 169-183, wherein the first vector, second vector, third vector, or any combination thereof, is a plasmid vector or a viral vector.
185. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) an N-terminal fragment of a mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a central fragment of the mScarlet gene, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a C-terminal fragment of the mScarlet gene and (ii) a nucleotide sequence encoding a third molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of the protein fragment encoded by N-terminal fragment of the mScarlet gene to a protein fragment encoded by the central fragment of the mScarlet gene, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of the protein fragment encoded by the central fragment of the mScarlet gene to the protein fragment encoded by the C-terminal fragment of the mScarlet gene, to produce a full-length mScarlet protein.
186. The method of paragraph 185, wherein the first vector encodes the sequence identified by SEQ ID NO: 121, the second vector encodes the sequence identified by SEQ ID NO: 123, and the third vector encodes the sequence identified by SEQ ID NO: 125.
187. A eukaryotic cell comprising: (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the fluorescent protein and (ii) a nucleotide sequence encoding a third molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the fluorescent protein to the central fragment of the fluorescent protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein, to produce a full-length fluorescent protein.
188. The eukaryotic cell of paragraph 187 wherein the eukaryotic cells are mammalian cells.
189. The eukaryotic cell of paragraph 187 or 188, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mScarlet, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP.
190. The eukaryotic cell of paragraph 189, wherein the fluorescent protein is mScarlet.
191. The eukaryotic cell of any one of paragraphs 187- 190, wherein the first intein is a split intein.
192. The eukaryotic cell of any one of paragraphs 185- 191, wherein the second intein is a split intein.
193. The eukaryotic cell of paragraph 191 or 192, wherein the split intein is a natural split intein.
194. The eukaryotic cell of paragraph 193, wherein the natural split intein is selected from DnaE inteins.
195. The eukaryotic cell of paragraph 194, wherein the DnaE inteins are selected from
Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpuDnaE) inteins.
196. The eukaryotic cell of paragraph 195, wherein the first intein is an NpaDnaE intein and the second intein is an NpaDnaE intein.
197. The eukaryotic cell of any one of paragraphs 185- 196, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
198. The eukaryotic cell of any one of paragraphs 185- 196, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
199. The eukaryotic cell of paragraph 198, wherein the non-coding RNA is a microRNA
(miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
200. The eukaryotic cell of any one of paragraphs 185- 199, wherein the first vector, second vector, third vector, or any combination thereof, is a plasmid vector or a viral vector.
201. A composition comprising the eukaryotic cell of any one of paragraph 185-200.
202. A kit comprising:
(a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein,
(b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the fluorescent protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and
(c) a third vector comprising a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the fluorescent protein,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the fluorescent protein to the central fragment of the fluorescent protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the fluorescent protein to the C-terminal fragment of the fluorescent protein, to produce a full-length fluorescent protein.
203. The kit of paragraph 202, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald,
Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mScarlet, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP. 204. The kit of paragraph 203, wherein the fluorescent protein is mScarlet.
205. The kit of any one of paragraphs 202-204, wherein the first intein is a split intein.
206. The kit of any one of paragraphs 202-205, wherein the second intein is a split intein.
207. The kit of paragraph 206, wherein the split intein is a natural split intein.
208. The kit of paragraph 207, wherein the natural split intein is selected from DnaE inteins.
209. The kit of paragraph 208, wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
210. The kit of paragraph 209, wherein the first intein is an NpaDnaE intein and the second intein is an NpaDnaE intein.
211. The kit of any one of paragraphs 202-210, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a protein.
212. The kit of any one of paragraphs 202-210, wherein the first molecule of interest, second molecule of interest, third molecule of interest, or any combination thereof is a non-coding ribonucleic acid (RNA).
213. The kit of paragraph 212, wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
214. The kit of any one of paragraphs 202-213, wherein the first vector, second vector, third vector, or any combination thereof, is a plasmid vector or a viral vector.
215. The kit of any one of paragraphs 202-214, further comprising any one or more of the following components: buffers, salts, cloning enzymes, competent cells, transfection reagents, antibiotics, and/or instructions for performing the methods described herein.
216. A transgenic selection method comprising delivering to a composition comprising eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding a first selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) upstream from a nucleotide sequence encoding an N-terminal intein protein fragment and (ii) a nucleotide sequence encoding a first molecule, and (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal intein protein fragment upstream from a second selectable marker protein fragment (e.g., antibiotic resistance protein fragment or fluorescent protein fragment) and (ii) a nucleotide sequence encoding a second molecule, wherein the N- terminal intein protein fragment and the C-terminal intein protein fragment catalyze joining of the first selectable marker protein fragment to the second selectable marker protein fragment to produce a full-length selectable marker protein.
217. A transgenic selection method comprising delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the central fragment of the selectable markerprotein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the selectable markerprotein to the C-terminal fragment of the selectable markerprotein, to produce a full-length selectable markerprotein.
218. A transgenic selection method comprising delivering to eukaryotic cells (a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein (e.g., antibiotic resistance protein or fluorescent protein), which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest, (b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a first central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, (c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a second central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a third intein and (ii) a nucleotide sequence encoding a third molecule of interest, and (d) a fourth vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the third intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the first central fragment of the selectable marker protein, the N-terminal fragment and the C- terminal fragment of the second intein catalyze joining of first central fragment of the selectable marke rprotein to the second central fragment of the selectable marker protein, the N-terminal fragment and the C-terminal fragment of the third intein catalyze joining of second central fragment of the selectable marker protein to the C-terminal fragment of the selectable marker protein to produce a full-length selectable marker protein.
219. The method of any one of paragraphs 216-218 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
220. The method of paragraph 219 further comprising selecting the transgenic eukaryotic cells that comprise the full-length selectable marker protein.
221. The method of any one of paragraphs 216-220, wherein the eukaryotic cells are mammalian cells.
222. The method of any one of paragraphs 216-221, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
223. The method of any one of paragraphs 216-222, wherein the intein is a split intein.
224. The method of paragraph 223, wherein the split intein is a natural split intein.
225. The method of paragraph 224, wherein the natural split intein is selected from DnaE inteins. 226. The method of paragraph 225, wherein the DnaE inteins are selected from Synechocystis sp.
DnaE (SspOnaE) inteins and Nostoc punctiforme (NpaDnaE) inteins.
227. The method of paragraph 223, wherein the split intein is an engineered split intein.
228. The method of paragraph 2278, wherein the engineered split intein is engineered from DnaB inteins.
229. The method of paragraph 228, wherein the engineered split intein is a SspOna S I intein.
230. The method of paragraph 229, wherein the engineered split intein is engineered from GyrB inteins.
231. The method of paragraph 230, wherein the engineered split intein is a SspGyrB S 11 intein.
232. The method of any one of paragraphs 216-231, wherein the molecules are selected from proteins.
233. The method of any one of paragraphs 216-231, wherein the molecules are selected from non-coding ribonucleic acids (RNAs).
234. The method of paragraph 233, wherein the non-coding RNAs are selected from microRNAs (miRNAs), antisense RNAs, short-interfering RNAs (siRNAs), and short-hairpin RNAs (shRNAs). 235. The method of any one of paragraphs 216-234, wherein the vectors are selected from plasmid vectors and viral vectors.
EXAMPLES
The present disclosure is further illustrated by the following Examples. These Examples are provided to aid in the understanding of the disclosure, and should not be construed as a limitation thereof.
Example 1. Antiobiotic Resistance Markers
Selectable markers are often used in genetic engineering to isolate cells with desired genotypes [1]. However, there are a limited number of well-characterized antibiotic resistance genes for use in eukaryotic cells and a limited number of fluorescent proteins whose spectra can be unambiguously differentiated by equipment in ordinary laboratories. Researchers often run into the problem of not having enough choices of selectable markers if they are to incorporate multiple transgenes into a cell. On the other hand, selection with multiple antibiotics at the same time is often harsh to cells. "Selectable marker recycling" may provide a work-around, however, requiring multiple rounds of transgenesis, selection and removal of selection markers [2]. To allow multiple transgenes to be selected by one selection scheme at the same time, we have created split antibiotics resistance and fluorescent protein genes wherein a gene encoding an antibiotic resistance or fluorescent protein is split into two or more segments fused to inteins ("markertrons") that can be rejoined by protein trans-splicing [3] (FIG. 1A). Each markertron is inserted onto a transgenic vector carrying a specific transgene. Delivery of transgenic vectors containing a set of markertrons yield cells harboring a subset or a complete set of the marketrons. Only cells containing a complete set of markertrons produce a fully reconstituted marker protein via protein splicing and thus passes through selection while cells with partial sets of markertrons are eliminated, achieving co-selection of cells containing all intended transgenes.
We started out with engineering 2-markertron intein-split resistance (Intres) genes for double transgenesis. Since flanking residues and local protein folding can affect efficiency of intein- mediated trans- splicing, we set out to identify split points in each of the four commonly used antibiotic resistance genes compatible with two well-characterized split inteins derived from
NpuDnaE [4, 5] and SspDnaB [6]. To facilitate assessment of the effectiveness of double transgenic selection, we cloned markertrons onto lentiviral vectors expressing TagBFP2 or mCherry fluorescent proteins as test transgenes (FIG. IB). Viral preparations were transduced into U20S cells which were then split into replicate plates with non-selective or selective media. Following appropriate passages for antibiotics selection, the two cell cultures were analyzed by flow cytometry. For hygromycin (Hygro) resistance gene, one "native" SspDnaB split point (G200:S201) with flanking residues "GS" and one "native" NpuDnaE split point (Y89:C90) with "YC" residues were tested. Both enabled successful selection when both N- and C-markertrons were transduced yielding >99% BFP+ mCherry+ double transgenic cells in selected cultures compared to <10% double-positive cells in non-selected culture (FIG. 3; Plasmid pairs 3,4 and 5,6). Cells transduced with either of the two markertrons did not survive hygromycin selection. In contrast, double transgenesis with conventional full-length non-split hygromycin vectors only allow for -20% enrichment of BFP+ mCherry+ cells (Plasmid pairs 97,98). We screened three addition potential split points (52S:53C),(240A:241C), and (292R:293C) for NpuDnaE with the obligatory cysteine residue on the C-extein junction and a residue on the N-extein junction that supported substantial trans-splicing activities in a previous report 7. We also incorporated six additional NpuDnaE split points by inserting an "artificial" cysteine on the C-extein junction to support splicing at ectopic sites yielding additional split points. In total, eight out of eleven split points tested supported hygromycin selection (FIG. 3). Similarly, for puromycin (Puro) (FIG. 4), neomycin (Neo) (FIG. 5) and blasticidin (Blast) (FIG. 6) resistance genes, we identified four, two, and one functional Intres pair(s), respectively. In all of these cases, cells transduced with either markertrons did not survive selection, while cells transduced with both yielded >95% double transgenic cells in selective cultures compared to <50% in non-selective cultures with the exception of Blasticidin(102) Intres, achieving lower but still significant enrichment of 91% double transgenic cells (FIGS. 3-6). Details of the split points of Intres genes and plasmids are presented in FIGS. 2A-2D and Table 1.
Table 1. Plasmids
Plasmid #* Plasmid Name Markertron
3 pLX-Hygro(1 -89)-NpuDnaE(N)-IRES-TagBFP2 Hygro(1-89)-NpuDnaE(N)
4 pLX-NpuDnaE(C)-Hygro(90-341)-IRES-mCherry NpuDnaE(C)-Hygro(90-341)
5 pLX-Hygro(1 -200)-SspDnaB(N)-IRES-TagBFP2 Hygro(1-200)-SspDnaB(N)
6 pLX-SspDnaB(C)-Hygro(201-341)-IRES-mCherry SspDnaB(C)-Hygro(201 -341)
7 pLX-Hygro(1 -52)-NpuDnaE(N)-IRES-TagBFP2 Hygro(1-52)-NpuDnaE(N)
8 pLX-NpuDnaE(C)-Hygro(53-341)-IRES-mCherry NpuDnaE(C)-Hygro(53-341)
9 pLX-Hygro(1 -240)-NpuDnaE(N)-IRES-TagBFP2 Hygro(1-240)-NpuDnaE(N)
10 pLX-NpuDnaE(C)-Hygro(241-341)-IRES-mCherry NpuDnaE(C)-Hygro(241 -341)
11 pLX-Hygro(1 -292)-NpuDnaE(N)-IRES-TagBFP2 Hygro(1-292)-NpuDnaE(N)
12 pLX-NpuDnaE(C)-Hygro(293-341)-IRES-mCherry NpuDnaE(C)-Hygro(293-341)
13 pLX-Blast(1-102)-NpuDnaE(N)-IRES-TagBFP2 Blast(1 -102)-NpuDnaE(N)
14 pLX-NpuDnaE(C)-Blast(103-140)-IRES-mCherry NpuDnaE(C)-Blast(103-140)
17 pLX-Puro(1 -119)-NpuDnaE(N)-IRES-TagBFP2 Puro(1-119)-NpuDnaE(N)
18 pLX-NpuDnaE(C)-Puro(insCys; 120-199)-IRES-mCherry NpuDnaE(C)-Puro(insCys; 120-199)
19 pLX-Puro(1 -100)-SspDnaB(N-S0)-IRES-TagBFP2 Puro(1-100)-SspDnaB(N-S0)
20 pLX-SspDnaB(C-S0)-Puro(101-199)-IRES-mCherry SspDnaB(C-S0)-Puro(101-199)
21 pLX-Neo(1-133)-NpuDnaE(N)-IRES-TagBFP2 Neo(1 -133)-NpuDnaE(N)
22 pLX-NpuDnaE(C)-Neo(134-267)-IRES-mCherry NpuDnaE(C)-Neo(134-267)
23 pLX-Neo(1-194)-NpuDnaE(N)-IRES-TagBFP2 Neo(1 -194)-NpuDnaE(N)
24 pLX-NpuDnaE(C)-Neo(195-267)-IRES-mCherry NpuDnaE(C)-Neo(195-267)
25 pLX-NpuDnaE(C)_Hygro(53-89)-NpuDnaE(N)-IRES-GFP NpuDnaE(C)_Hygro(53-89)-NpuDnaE(N)
26 pLX-NpuDnaE(C)_Hygro(53-239)-NpuDnaE(N)-IRES-GFP NpuDnaE(C)_Hygro(53-239)-NpuDnaE(N)
27 pCR8-Bsal->ccdbCam<-Bsal-NpuDnaE(N)-MD1-68-15
28 pCR8-NpuDnaE(C)_Bsal->ccdbCam<-Bsal-MD1-68-18
29 pCR8-Bsal->ccdbCam<-Bsal-SspDnaE(N)-MD1-68-12
30 pCR8-SspDnaE(C)_Bsal->ccdbCam<-Bsal-MD1-68-13
31 pCR8-Bsal->ccdbCam<-Bsal-SspDnaB(N-S0)-25-135-18
32 pCR8-SspDnaB(C-S0)_Bsal->ccdbCam<-Bsal-25-155-41
Figure imgf000051_0001
Figure imgf000052_0001
Example 2. Gateway- Compatible Lentiviral Vectors
To facilitate adoption of Intres markers, we created Gateway-compatible lentiviral vectors for convenient restriction-ligation-independent LR clonase recombination of transgenes 8 (FIG. 7A). We tested the functionality of these vectors by recombining TagBPF2 and mCherry, respectively to the N- and C-Intres vectors and found robust selection of double transgenic cells (FIG. 7B). One potential utility of Intres vectors is to install different fluorescent markers in cells to label different cellular compartments. To explore such utility, we cloned in NLS-GFP and LifeAct- mScarlet 9, which label nucleus and F-actin, respectively, by Gateway recombination to
conventional full length (FL) non-split hygromycin selectable vectors or 2-markertron hygromycin Intres vectors and transduced cells with either sets of plasmids, followed by antibiotic selection (FIG. 7C). The sample transduced with non-split selectable plasmids contained both singly and doubly labelled cells, while cell transduced with Intres plasmids were all doubly labelled (FIG. 7C).
Example 3. Fluorescent Markers
To test whether split fluorescent markers can be used for transgene selection, we screened for NpuDnaE split points for mScarlet fluorescent protein (FIG. 8A) and identified four split points allowing for >96% enrichment of double transgenic cells and three other split points enabling >60% enrichment of double transgenic cells in mScarlet-gated population, compared to <20% double transgenic cells in non-gated population (FIG. 8B).
Example 4. Higher Degree Split Markers
With the split points identified for 2-markertron Intres genes, we set out to engineer higher degree split markers. We tested combinations of splits points to partition a marker gene into three or more markertrons to allow for co-selection of more than two "unlinked" transgenes with one antibiotics (FIGS. 9A-9B). To identify pairs of split points that would allow such "Intres chain", we cloned 3-split markertrons into three lentiviral vectors each carrying one of three fluorescent transgenes TagBFP2, EGFP, or mCherry, that will allow us to assess effectiveness of selection by flow cytometry (FIG. 9C). Since hygromycin resistance gene is the longest and provides the most split points for testing, we focused on engineering 3-markertron hygromycin Intres. We tested two 3-markertron hygromycin Intres using two intervening NpuDnaE inteins, two using NpuDnaE for the first intein and SspDnaB for the second intein, as well as two using SspDnaB for the first intein and NpuDnaE for the second intein (FIG. 9D). Five of these six 3-markertron hygromycin Intres enabled >97% and with the remaining one enabling 80% triple transgenic selection in hygromycin- selected cultures compared to <15% triple transgenic cells in non- selected cultures. Samples with leave-one-out transduction did not yield any viable cells after hygromycin selection while cells transduced with non-split hygromycin vectors yielded only 7% triple transgenic cells after selection.
To facilitate the use of 3-markertron Intres, we created Gateway compatible lentiviral vectors with these markers (FIG. 10A). Three sets of these vectors were each tested by recombining TagBFP (as transgene 1), EGFP (as transgene 2) and mCherry (as transgene 3) into the N-, M-, and C-Intres Gateway destination vectors and used to transduce U20S cells, which were then split and cultured in hygromycin selection or non-selective media (FIG. 10B). Two weeks after selection, cells were analyzed by flow cytometry. All three sets of 3-markertron hygromycin Intres plasmids support triple transgenic cell selection of >99% compared to <25% in the non-selected cultures (FIG. IOC).
We further tested the feasibility of 4-markertron hygromycin Intres genes (FIG. 11). Here, we used an enhanced variant of NpuDnaE intein known as NpuDnaGEP 10 fused with leucine zipper motifs 11 in combination with the SspDnaB intein. While transduction of all four plasmids containing constituent markertrons produced cells that survived hygromycin selection, leave-one- out transduction did not yield any survival (Table 2).
Table 2. Survival of cells transduced with ("+") or without ("-") lentiviruses prepared from the indicated plasmids.
Figure imgf000054_0001
Example 5. Biallelic Knock-In at the AAVS1 Locus
CRISPPv/Cas has recently emerged as a powerful technology for genome engineering and editing. Although gene knockout based on NHEJ-mediated insertions/deletions (indels) occur at high frequency, precise editing and knock-in based on homology directed repair (HDR) using exogenous repair templates (a.k.a targeting constructs) are inefficient. We tested whether split selectable markers can be used to enrich for cells with biallelic knock- in at the AAVS 1 locus. We constructed targeting constructs with homology arms flanking the target site, and splice acceptor-2A peptide to trap the markertrons within intron one of the host gene PPP1R12C. However, we did not obtain any live cells after CRISPR/Cas knock-in experiments using these targeting constructs and two weeks of antibiotic selection (data not shown). We suspected that the endogenous promoter of the host gene PPP1R12C might not drive sufficient expression of markertrons to reconstitute enough antibiotic resistance protein to counteract actions of the antibiotics. We thus tested an alternative strategy to express Intres markertrons by TetO promoter whose activity can be titrated by doxycycline (dox) concentration. To allow comparison of Intres-mediated biallelic selection versus full-length (FL) non-split selectable markers, we implemented several different targeting construct designs. First, we drive expression of a full-length (FL) resistance gene (e.g., Hygro) together with rtTA under a constitutive EFla promoter and a separate test Intres (e.g., Blast Intres) under a dox-inducible TetO promoter (FIG. 12B, Plasmids 109 and 110). This allow comparison of full length and split selectable markers within the same constructs. To allow fair comparison of full length versus split markers driven by the same TetO promoter, we constructed two similar plasmids 107 and 108 (cf. Plasmids 109 and 110), wherein full-length antibiotic resistance gene (Blast) is placed downstream of the TetO promoter (FIG. 12A). To enable single-cell quantification of biallelic targeting and to demonstrate the feasibility of incorporating two transgenes into two AAVS 1 alleles, we appended EGFP and mScarlet fluorescent genes downstream of the test split or non-split markers via self-cleaving 2A peptide. Similarly, to test Hygro Intres, we swapped the
EFla and TetO-driven markers so that FL Hygro or Hygro Intres were placed downstream of TetO and FL Blast downstream of EFla (FIGS. 12C-12D; Plasmids 111-114). We co-transfected pX330- AAVS 1 (Plasmid 106) containing Cas9 and sgRNA targeting AAVS 1, and the different pairs of targeting constructs to HEK293T cells, split into triplicate doxycycline-containing media without antibiotics, with blasticidin, or with hygromycin at the subsequent passages. Two weeks after selection, we analyzed the cultures for biallelic targeting by flow cytometric measurement of GFP and RFP fluorescence (FIG. 12E). As expected, non- selected cultures harbored small fraction (<1%) of biallelic knock-in GFP+/RFP+ cells (FIG. 12E; Selection = None). Selection of antibiotics where corresponding FL antibiotic resistance genes were present on targeting constructs yielded < 30% biallelic knock-in cells (FIG. 12E; Blast: TC a,c,d; Hygro: TC a,b,c). In contrast, selection of antibiotics where corresponding Intres are present on the targeting constructs yielded 75% (Fig 6e; Blast Intres: TC b) and 88% (Fig 6e; Hygro Intres: TC d) biallelic knock-in cells.
In the Examples above, we have engineered split antibiotic resistance and fluorescent protein genes that can allow selection for two or more "unlinked" transgenes. By inserting unnatural residues at selectable markers, we showed that novel high-efficiency split points can be utilized, expanding the positions available for engineering. We demonstrated that split selectable markers can be incorporated into lentiviral vectors or gene targeting constructs in CRISPR/Cas9 genome editing experiments to enable enrichment of cells with double transgenesis or biallelic knock-ins. By combining two or more splits points, we showed that 3- and 4-split markers can be generated to allow higher degree transgenic selection. Future development of even higher-degree split selectable markers may enable "hyper-engineering" of cells containing tens of transgenes or targeted knock- ins. Materials and Methods
Cloning
To generate a test plasmid for each markertron, we first generated a Gateway donor plasmid containing its ORF and then recombine into lentiviral destination vector with TagBFP2 (Plasmid 94: pLX-DEST-IRES-TagBFP2), EGFP (Plasmid 95: pLX-DEST-IRES-EGFP), or mCherry (Plasmid 96: pLX-DEST-IRES-mCherry) reporters, which were derived from pLX302
(addgene.org/25896/) by removing Puromycin resistance gene and inserting IRES -fluorescent genes downstream of the Gateway cassette. The markertron-ORF Gateway donor plasmids were generated either by a nested fusion PCR procedure to combine intein with the coding sequence of fragments of the selectable marker followed by insertion into the pCR8-GW-TOPO plasmid by sequence- and ligation-independent cloning (SLIC) (Li, M.Z. & Elledge, S.J. SLIC: a method for sequence-and ligation-independent cloning. Gene Synthesis: Methods and Protocols, 51-59 (2012)), or PCR- amplifying the relevant fragment of the selectable marker followed by insertion into "scaffold" plasmids (Plasmids 27-32) containing the intein sequences by SLIC. DNA sequences encoding inteins were codon optimized for Homo sapiens, and synthesized as GBlock (IDT), with
AC 1947GB encoding NpuDnaE intein, AC 1949GB encoding SspDnaB intein. Selectable marker fragments were amplified from plasmids containing these markers. See Table 1 for plasmids.
Cell Culture
All cells were cultivated in Dulbecco's modified Eagle's medium (DMEM) (Sigma) with 10% fetal bovine serum (FBS)(Lonza), 4% Glutamax (Gibco), 1% Sodium Pyruvate (Gibco) and penicillin-streptomycin (Gibco). Incubator conditions were 37°C and 5% CO2.
Virus Production
A viral packaging mix of pLPl, pLP2, and VSV-G were co-transfected with each lentiviral vector into Lenti-X 293T cells (ClonTech), seeded the day before in 6-well plates at a concentration of 1.2xl06 cells per well, using Lipofectamine 3000. Media was changed 6h after transfection then incubated overnight. 28 hour post transfection, the media supernatant containing virus was filtered using 45uM PES filters then stored at -80°C until use.
Transduction
The day prior to transduction, target cells (HEK293T, MCF7, U2-OS) were seeded into 12- well plates at a density of 1.5xl05 cells per well. Prior to transduction, media was changed to media containing 10 μg/mL polybrene, 1 mL per well. 250 μΐ^ of each respective virus (500 μΐ^ total for experimental samples with two viruses added) was added to each well and incubated overnight. Media was changed 24 hour post infection. 4 day post infection cells were split into duplicate plates. 5 day post infection media with antibiotic (hygromycin) was added to each respective well of one replicate plate (the other remained under no selection). Antibiotic selection continued for 2 weeks before analysis on FACS.
Fluorescent- Activated Cell Sorting
Cells were trypisinized, suspended in media then analyzed on a LSRFortessa X-20 (BD Bioscience) flow cytometer using FACSDiVa software, version 8, on an HP Z230 workstation. Fifty thousand events were collected each run.
CONSTRUCTS AND SEQUENCES
NpuDnaE(N)
CLSYETEILTVEYGLLPIGKIVEKRIECTVYSVDNNGNIYTQPVAQWHDRGEQEVFEYCLEDGSLIRATKDHKF MTVDGQMLPIDEIFERELDLMRVDNLPN (SEQ ID NO: 7)
NpuDnaE(C)
IKIATRKYLGKQNVYDIGVERDHNFALKNGFIASN (SEQ ID NO: 8)
SspDnaB(N-SO)
CISGDSLISLASTGKRVSIKDLLDEKDFEIWAINEQTMKLESAKVSRVFCTGKKLVYILKTRLGRTIKATANHRF LTIDG WKRLDELS LKEHI ALPRKLES S S LQL (SEQ ID NO: 9)
SspDnaB(C-SO)
SPEIEKLSQSDIYWDSIVSITETGVEEVFDLTVPGPHNFVANDIIVHN (SEQ ID NO: 10)
NpuDnaE(N)-LZA
CLSYETEILTVEYGLLPIGKIVEKRIECTVYSVDNNGNIYTQPVAQWHDRGEQEVFEYCLEDGSLIRATKDHKF MTVDGQMLPIDEIFERELDLMRVDNLPNGGGGSGSAQLEKELQALEKKLAQLEWENQALEKELAQ (SEQ ID NO: 11)
LZB-NpuDnaGEP(C)
AQLKKKLQANKKELAQLKWKLQALKKKLAQGGGGSGSMIKIATRKYLGKQNVYDIGVGEPHNFALKNGFIA SN (SEQ ID NO: 12)
NpuDnaGFP(C)
IKIATRKYLGKQNVYDIGVGEPHNFALKNGFIASN (SEQ ID NO: 13)
LZA
AQLEKELQALEKKLAQLEWENQALEKELAQ (SEQ ID NO: 14)
LZB
AQLKKKLQANKKELAQLKWKLQALKKKLAQ (SEQ ID NO: 15) SspDnaE(N)
CLSFGTEILTVEYGPLPIGKIVSEEINCSVYSVDPEGRVYTQAIAQWHDRGEQEVLEYELEDGSVIRATSDHRFL TTDYQLLAIEEIFARQLDLLTLENIKQTEEALDNHRLPFPLLDAGTIK (SEQ ID NO: 16) SspDnaE(C)
VKVIGRRSLGVQRIFDIGLPQDHNFLLANGAIAAN (SEQ ID NO: 17)
SspDnaB(N)
CISGDSLISLA (SEQ ID NO: 18)
SspDnaB(C)
STGKRVSIKDLLDEKDFEIWAINEQTMKLESAKVSRVFCTGKKLVYILKTRLGRTIKATANHRFLTIDGWKRLD ELSLKEHIALPRKLESSSLQLSPEIEKLSQSDIYWDSIVSITETGVEEVFDLTVPGPHNFVANDIIVHN (SEQ ID NO: 19)
Plasmid 3: pLX-Hygro(l-89)-NpuDnaE(N)-IRES-TagBFP2
Protein = Hygro(l-89)-NpuDnaE(N)
Vector sequence (SEQ ID NO:20)
Amino acid sequence (SEQ ID NO:21)
Plasmid 4: pLX-NpuDnaE(C)-Hygro(90-341)-IRES-mCherry
Protein = NpuDnaE(C)-Hygro(90-341)
Vector sequence (SEQ ID NO:22)
Amino acid sequence (SEQ ID NO:23)
Plasmid 5: pLX-Hygro(l-200)-SspDnaB(N)-IRES-TagBFP2
Protein = Hygro(l-200)-SspDnaB(N)
Vector sequence (SEQ ID NO:24)
Amino acid sequence (SEQ ID NO:25)
Plasmid 6: pLX-SspDnaB(C)-Hygro(201-341)-IRES-mCherry
Protein = SspDnaB(C)-Hygro(201-341)
Vector sequence (SEQ ID NO:26)
Amino acid sequence (SEQ ID NO:27)
Plasmid 7: pLX-Hygro(l-52)-NpuDnaE(N)-IRES-TagBFP2
Protein = Hygro(l-52)-NpuDnaE(N)
Vector sequence (SEQ ID NO:28)
Amino acid sequence (SEQ ID NO:29) Plasmid 8: pLX-NpuDnaE(C)-Hygro(53-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(53-341)
Vector sequence (SEQ ID NO: 30)
Amino acid sequence (SEQ ID NO:31)
Plasmid 9: pLX-Hygro(l -240)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro( l-240)-NpuDnaE(N)
Vector sequence (SEQ ID NO:32)
Amino acid sequence (SEQ ID NO:33)
Plasmid 10: pLX-NpuDnaE(C)-Hygro(241-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(241-341)
Vector sequence (SEQ ID NO: 34)
Amino acid sequence (SEQ ID NO:35)
Plasmid 11: pLX-Hygro( l-292)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro(l-292)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 36)
Amino acid sequence (SEQ ID NO: 37)
Plasmid 12: pLX-NpuDnaE(C)-Hygro(293-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(293-341)
Vector sequence (SEQ ID NO:38)
Amino acid sequence (SEQ ID NO: 39)
Plasmid 13: pLX-Blast(l -102)-NpuDnaE(N)-IRES-TagBFP2 Protein = Blast(l-102)-NpuDnaE(N)
Vector sequence (SEQ ID NO:40)
Amino acid sequence (SEQ ID NO:41)
Plasmid 14: pLX-NpuDnaE(C)-Blast(103-140)-IRES-mCherry Protein = NpuDnaE(C)-Blast(103-140)
Vector sequence (SEQ ID NO:42)
Amino acid sequence (SEQ ID NO:43) Plasmid 17: pLX-Puro( l- l 19)-NpuDnaE(N)-IRES-TagBFP2 Protein = Puro(l-l 19)-NpuDnaE(N)
Vector sequence (SEQ ID NO:44)
Amino acid sequence (SEQ ID NO:45) Plasmid 18: pLX-NpuDnaE(C)-Pm-o(insCys; 120-199)-IRES-mCherry Protein = NpuDnaE(C)-Puro(insCys; 120-199)
Vector sequence (SEQ ID NO:46)
Amino acid sequence (SEQ ID NO:47) Plasmid 19: pLX-Puro( l -100)-SspDnaB(N-S0)-IRES-TagBFP2 Protein = Puro(l- 100)-SspDnaB(N-S0)
Vector sequence (SEQ ID NO:48)
Amino acid sequence (SEQ ID NO:49) Plasmid 20: pLX-SspDnaB(C-S0)-Puro(101 -199)-IRES-mCherry Protein = SspDnaB(C-S0)-Puro(101 -199)
Vector sequence (SEQ ID NO:50)
Amino acid sequence (SEQ ID NO:51) Plasmid 21: pLX-Neo(l-133)-NpuDnaE(N)-IRES-TagBFP2
Protein = Neo(l -133)-NpuDnaE(N)
Vector sequence (SEQ ID NO:52)
Amino acid sequence (SEQ ID NO:53) Plasmid 22: pLX-NpuDnaE(C)-Neo(134-267)-IRES-mCherry
Protein = NpuDnaE(C)-Neo(134-267)
Vector sequence (SEQ ID NO:54)
Amino acid sequence (SEQ ID NO:55) Plasmid 23 : pLX-Neo( 1 - 194)-NpuDnaE(N)-IRES -TagBFP2
Protein = Neo(l- 194)-NpuDnaE(N)
Vector sequence (SEQ ID NO:56)
Amino acid sequence (SEQ ID NO:57)
Plasmid 24: pLX-NpuDnaE(C)-Neo( 195-267)-IRES-mCherry
Protein = NpuDnaE(C)-Neo(195-267)
Vector sequence (SEQ ID NO:58)
Amino acid sequence (SEQ ID NO:59)
Plasmid 25: pLX-NpuDnaE(C)_Hygro(53-89)-NpuDnaE(N)-IRES-GFP Protein = NpuDnaE(C)_Hygro(53-89)-NpuDnaE(N)
Vector sequence (SEQ ID NO:60)
Amino acid sequence (SEQ ID NO:61)
Plasmid 26: pLX-NpuDnaE(C)_Hygro(53-239)-NpuDnaE(N)-IRES-GFP Protein = NpuDnaE(C)_Hygro(53-239)-NpuDnaE(N) Vector sequence (SEQ ID NO:62)
Amino acid sequence (SEQ ID NO:63)
Plasmid 27: pCR8-BsaI->ccdbCam<-BsaI-NpuDnaE(N)-MDl-68-15 (SEQ ID NO:64)
Plasmid 28: pCR8-NpuDnaE(C)_BsaI->ccdbCam<-BsaI-MDl-68-18 (SEQ ID NO:65) Plasmid 29: pCR8-BsaI->ccdbCam<-BsaI-SspDnaE(N)-MDl-68-12 (SEQ ID NO:66) Plasmid 30: pCR8-SspDnaE(C)_BsaI->ccdbCam<-BsaI-MDl-68-13 (SEQ ID NO:67) Plasmid 31: pCR8-BsaI->ccdbCam<-BsaI-SspDnaB(N-S0)-25-135-18 (SEQ ID NO:68) Plasmid 32: pCR8-SspDnaB(C-S0)_BsaI->ccdbCam<-BsaI-25-155-41 (SEQ ID NO:69)
Plasmid 33: pLX-mScarlet(l-46)-NpuDnaE(N)_LZA-IRES-TagBFP2
Protein = mScarlet(l-46)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO:70)
Amino acid sequence (SEQ ID NO:71)
Plasmid 34: pLX-LZB_NpuDnaE(C)-mScarlet(insCys;47-232)-IRES-TagBFP2 Protein = LZB_NpuDnaE(C)-mScarlet(insCys;47-232)
Vector sequence (SEQ ID NO:72)
Amino acid sequence (SEQ ID NO:73)
Plasmid 35: pLX-mScarlet(l-48)-NpuDnaE(N)_LZA-IRES-TagBFP2
Protein = mScarlet(l-48)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO:74)
Amino acid sequence (SEQ ID NO:75)
Plasmid 36: pLX-LZB_NpuDnaE(C)-mScarlet(insCys;49-232)-IRES-GFP
Protein = LZB_NpuDnaE(C)-mScarlet(insCys;49-232)
Vector sequence (SEQ ID NO:76)
Amino acid sequence (SEQ ID NO:77)
Plasmid 37: pLX-mScarlet(l-51)-NpuDnaE(N)_LZA -IRES-TagBFP2
Protein = mScarlet(l-51)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO:78)
Amino acid sequence (SEQ ID NO:79)
Plasmid 38: pLX-LZB_NpuDnaE(C)-mScarlet(insCys;52-232)-IRES-GFP Protein = LZB_NpuDnaE(C)-mScarlet(insCys;52-232)
Vector sequence (SEQ ID NO: 80)
Amino acid sequence (SEQ ID NO:81) Plasmid 39: pLX-mScarlet( 1 -75)-NpuDnaE(N)_LZA-IRES -TagBFP2 Protein = mScarlet(l-75)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO: 82)
Amino acid sequence (SEQ ID NO:83) Plasmid 40: pLX-LZB_NpuDnaE(C)-mScarlet(insCys;76-232)-IRES-GFP Protein = LZB_NpuDnaE(C)-mScarlet(insCys;76-232)
Vector sequence (SEQ ID NO: 84)
Amino acid sequence (SEQ ID NO: 85) Plasmid 41: pLX-mScarlet(l-122)-NpuDnaE(N)_LZA-IRES-TagBFP2 Protein = mScarlet(l-122)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO: 86)
Amino acid sequence (SEQ ID NO: 87) Plasmid 42: pLX-LZB_NpuDnaE(C)-mScarlet(insCys; 123-232)-IRES-GFP Protein = LZB_NpuDnaE(C)-mScarlet(insCys; 123-232)
Vector sequence (SEQ ID NO:88)
Amino acid sequence (SEQ ID NO: 89)
Plasmid 43: pLX-mScarlet(l-140)-NpuDnaE(N)_LZA-IRES-TagBFP2 Protein = mScarlet(l-140)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO:90)
Amino acid sequence (SEQ ID NO:91)
Plasmid 44: pLX-LZB_NpuDnaE(C)-mScarlet(insCys; 141-232)-IRES-GFP Protein = LZB_NpuDnaE(C)-mScarlet(insCys; 141-232)
Vector sequence (SEQ ID NO:92)
Amino acid sequence (SEQ ID NO:93)
Plasmid 45: pLX-mScarlet(l-163)-NpuDnaE(N)_LZA-IRES-TagBFP2 Protein = mScarlet(l-163)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO:94)
Amino acid sequence (SEQ ID NO:95)
Plasmid 46: pLX-LZB_NpuDnaE(C)-mScarlet(insCys; 164-232)-IRES-GFP Protein = LZB_NpuDnaE(C)-mScarlet(insCys; 164-232) Vector sequence (SEQ ID NO:96)
Amino acid sequence (SEQ ID NO:97)
Plasmid 47: pCR8-TagBFP2
Protein = TagBFP2
Vector sequence (SEQ ID NO:98)
Amino acid sequence (SEQ ID NO:99)
Plasmid 48: pCR8-mCherry
Protein = mCherry
Vector sequence (SEQ ID NO: 100)
Amino acid sequence (SEQ ID NO: 101)
Plasmid 49: pLX-DEST-IRES-Hygro(l-89)-NpuDnaE(N) Protein = Hygro(l-89)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 102)
Amino acid sequence (SEQ ID NO: 103)
Plasmid 50: pLX-DEST-IRES-NpuDnaE(C)-Hygro(90-341) Protein = NpuDnaE(C)-Hygro(90-341)
Vector sequence (SEQ ID NO: 104)
Amino acid sequence (SEQ ID NO: 105)
Plasmid 51: pLX-[TagBFP2]-IRES-Hygro(l-89)-NpuDnaE(N) Vector sequence (SEQ ID NO: 106)
Plasmid 52: pLX-[mCherry]-IRES-NpuDnaE(C)-Hygro(90-341) Vector sequence (SEQ ID NO: 107) Plasmid 53: pLX-DEST-IRES-Puro(l-l 19)-NpuDnaE(N) Protein = Puro(l-l 19)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 108)
Amino acid sequence (SEQ ID NO: 109) Plasmid 54: pLX-DEST-IRES-NpuDnaE(C)-Puro(120-199) Protein = NpuDnaE(C)-Puro(120-199)
Vector sequence (SEQ ID NO: 110)
Amino acid sequence (SEQ ID NO: 111) Plasmid 55: pLX-[TagBFP2]-IRES-Puro(l-l 19)-NpuDnaE(N) Vector sequence (SEQ ID NO: 112) Plasmid 56: pLX-[mCherry]-IRES-NpuDnaE(C)-Puro(120-199)
Vector sequence (SEQ ID NO: 113)
Plasmid 57: pLX-DEST-IRES-Neo(l-194)-NpuDnaE(N)
Protein = Neo(l-194)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 114)
Amino acid sequence (SEQ ID NO: 115)
Plasmid 58: pLX-DEST-IRES-NpuDnaE(C)-Neo(195-267)
Protein = NpuDnaE(C)-Neo(195-267)
Vector sequence (SEQ ID NO: 116 )
Amino acid sequence (SEQ ID NO: 117)
Plasmid 59: pLX-[TagBFP2]-IRES-Neo(l-194)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 118)
Plasmid 60: pLX-[mCherry]-IRES-NpuDnaE(C)-Neo(195-267)
Vector sequence (SEQ ID NO: 119)
Plasmid 61: pLX-mScarlet(l-51)-NpuDnaE(N)-LZA-IRES-TagBFP2
Protein = mScarlet(l-51)-NpuDnaE(N)-LZA
Vector sequence (SEQ ID NO: 120)
Amino acid sequence (SEQ ID NO: 121)
Plasmid 62: pLX-LZB -NpuDnaE(C)-mScarlet(AC,52- 163)-NpuDnaE(N)_LZA-IRES -EGFP Protein = LZB-NpuDnaE(C)-mScarlet(AC;52-163)-NpuDnaE(N)_LZA
Vector sequence (SEQ ID NO: 122)
Amino acid sequence (SEQ ID NO: 123)
Plasmid 63: pLX-LZB-NpuDnaE(C)-mScarlet(AC; 164-232)-IRES-EGFP
Protein = LZB-NpuDnaE(C)-mScarlet(AC; 164-232)
Vector sequence (SEQ ID NO: 124)
Amino acid sequence (SEQ ID NO: 125)
Plasmid 64: pLX-Hygro(l-69)-NpuDnaE(N)-IRES-TagBFP2
Protein = Hygro( 1 -69)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 126)
Amino acid sequence (SEQ ID NO: 127)
Plasmid 65: pLX-NpuDnaE(C)-Hygro(AC;70-341)-IRES-mCherry
Protein = NpuDnaE(C)-Hygro(AC;70-341)
Vector sequence (SEQ ID NO: 128)
Amino acid sequence (SEQ ID NO: 129) Plasmid 66: pLX-Hygro(l-131)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro( l -131)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 130)
Amino acid sequence (SEQ ID NO: 131)
Plasmid 67: pLX-NpuDnaE(C)-Hygro(AC; 132-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(AC;132-341)
Vector sequence (SEQ ID NO: 132)
Amino acid sequence (SEQ ID NO: 133)
Plasmid 68: pLX-Hygro(l-171)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro( 1 - 17 l)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 134)
Amino acid sequence (SEQ ID NO: 135)
Plasmid 69: pLX-NpuDnaE(C)-Hygro(AC; 172-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(AC;172-341)
Vector sequence (SEQ ID NO: 136)
Amino acid sequence (SEQ ID NO: 137)
Plasmid 70: pLX-Hygro( l -218)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro( 1 -218)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 138)
Amino acid sequence (SEQ ID NO: 139)
Plasmid 71: pLX-NpuDnaE(C)-Hygro(AC;219-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(AC;219-341)
Vector sequence (SEQ ID NO: 140)
Amino acid sequence (SEQ ID NO: 141)
Plasmid 72: pLX-Hygro(l-259)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro( 1 -259)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 142)
Amino acid sequence (SEQ ID NO: 143)
Plasmid 73: pLX-NpuDnaE(C)-Hygro(AC;260-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(AC;260-341)
Vector sequence (SEQ ID NO: 144)
Amino acid sequence (SEQ ID NO: 145) Plasmid 74: pLX-Hygro(l-277)-NpuDnaE(N)-IRES-TagBFP2 Protein = Hygro(l -277)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 146)
Amino acid sequence (SEQ ID NO: 147)
Plasmid 75: pLX-NpuDnaE(C)-Hygro(AC; 278-341)-IRES-mCherry Protein = NpuDnaE(C)-Hygro(AC;278-341)
Vector sequence (SEQ ID NO: 148)
Amino acid sequence (SEQ ID NO: 149)
Plasmid 76: pLX-Puro(l-32)-NpuDnaE(N)-IRES-TagBFP2
Protein = Puro(l-32)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 150)
Amino acid sequence (SEQ ID NO: 151)
Plasmid 77: pLX-NpuDnaE(C)-Piiro(AC;33-199)-IRES-mCherry
Protein = NpuDnaE(C)-Puro(AC;33-199)
Vector sequence (SEQ ID NO: 152)
Amino acid sequence (SEQ ID NO: 153)
Plasmid 78: pLX-Puro( l -84)-NpuDnaE(N)-IRES-TagBFP2
Protein = Puro(l-84)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 154)
Amino acid sequence (SEQ ID NO: 155)
Plasmid 79: pLX-NpuDnaE(C)-Puro(AC;85-199)-IRES-mCherry
Protein = NpuDnaE(C)-Puro(AC;85-199)
Vector sequence (SEQ ID NO: 156)
Amino acid sequence (SEQ ID NO: 157)
Plasmid 80: pLX-Puro(l-137)-NpuDnaE(N)-IRES-TagBFP2
Protein = Puro(l -137)-NpuDnaE(N)
File = pLX-[PuroKC3(N)-NpuDnaE(N)-25-131 -29"]-IRES-TagBFP2-25-133-6 Vector sequence (SEQ ID NO: 158)
Amino acid sequence (SEQ ID NO: 159)
Plasmid 81: pLX-NpuDnaE(C)-Puro(AC; 138-199)-IRES-mCherry
Protein = NpuDnaE(C)-Puro(AC; 138-199)
Vector Sequence (SEQ ID NO: 160)
Amino acid sequence (SEQ ID NO: 161) Plasmid 82: pLX-Puro(l- 158)-NpuDnaE(N)-IRES-TagBFP2
Protein = Puro(l-158)-NpuDnaE(N)
Vector Sequence (SEQ ID NO: 162)
Amino acid sequence (SEQ ID NO: 163)
Plasmid 83: pLX-NpuDnaE(C)-Puro(AC; 159-199)-IRES-mCherry Protein = NpuDnaE(C)-Puro(AC; 159-199)
Vector Sequence (SEQ ID NO: 164)
Amino acid sequence (SEQ ID NO: 165)
Plasmid 84: pLX-Puro(l -180)-NpuDnaE(N)-IRES-TagBFP2
Protein = Puro(l-180)-NpuDnaE(N)
Vector Sequence (SEQ ID NO: 166)
Amino acid sequence (SEQ ID NO: 167)
Plasmid 85: pLX-NpuDnaE(C)-Puro(AC; 181-199)-IRES-mCherry Protein = NpuDnaE(C)-Puro(AC; 181-199)
Vector Sequence (SEQ ID NO: 168)
Amino acid sequence (SEQ ID NO: 169)
Plasmid 86: pLX-Blast( l-58)-NpuDnaE(N)-IRES-TagBFP2
Protein = Blast(l-58)-NpuDnaE(N)
Vector Sequence (SEQ ID NO: 170)
Amino acid sequence (SEQ ID NO: 171)
Plasmid 87: pLX-NpuDnaE(C)-BIast(59-140)-IRES-mCherry Protein = NpuDnaE(C)-Blast(59-140)
Vector Sequence (SEQ ID NO: 172)
Amino acid sequence (SEQ ID NO: 173)
Plasmid 88: pLX-NpuDnaE(C)-HygroBA-SspDoaB( -SO)-IRES-EGFP Protein = NpuDnaE(C)-Hy,£:ro(53-200)-SspDoaB(N-S0)
Vector Sequence (SEQ ID NO: 174)
Amino acid sequence (SEQ ID NO: 175)
Plasmid 89: pLX-SspDnaB(C-S0)-Hygro(201-341 )-IRES-mCherry Protein = SspDnaB(C-S0)-Hygro(20i-341)
Vector Sequence (SEQ ID NO: 176)
Amino acid sequence (SEQ ID NO: 177) Plasmid 90: pLX-NpuDiiaF C)-Hygro(90-2(K))-SspDnaB( T-S0)-II^S-EGFP Protein = NpuDoaE(C)-Hygro(90-200)-SspDnaB N-SO)
Vector Sequence (SEQ ID NO: 178)
Amino acid sequence (SEQ ID NO: 179)
Plasmid 91: pLX-Hygro(l-200)-SspDnaB(N-S0)-IRES-TagBFP2
Protein = Hygro(l-200)-SspDnaB(N-S0)
Vector sequence (SEQ ID NO: 180)
Amino acid sequence (SEQ ID NO: 181)
Plasmid 92: pLX-SspDnaB(C-S0)-Hygro(201 -240)-NpuDnaE( )-IRES-EGFP Protein = SspDnaB(C-S0)-Hygro(201-2 0)-NpuDnaE( )
Vector sequence (SEQ ID NO: 182)
Amino acid sequence (SEQ ID NO: 183)
Plasmid 93: pLX-SspDnaB(C-S0)-Hygro(201-292)-Npi;DnaE(N )-IRES-EGFP Protein = SspDnaB(C-S0)-Hygro(201-292)-NpuDnaE( )
Vector sequence (SEQ ID NO: 184)
Amino acid sequence (SEQ ID NO: 185)
Plasmid 94: pLX-DEST-IRES-TagBFP2 (SEQ ID NO: 186)
Plasmid 95: pLX-DEST-IRES-EGFP (SEQ ID NO: 187) Plasmid 96: pLX-DEST-IRES-mCherry (SEQ ID NO: 188)
Plasmid 97: pLX-Hygro-IRES-TagBFP2
Vector sequence (SEQ ID NO: 189) Plasmid 98: pLX-Hygro-IRES-mCherry
Vector sequence (SEQ ID NO: 190)
Plasmid 99: pLX-Puro-IRES-TagBFP2
Vector sequence (SEQ ID NO: 191)
Plasmid 100: pLX-Puro-IRES-mCherry
Vector sequence (SEQ ID NO: 192)
Plasmid 101: pLX-Hygro-IRES-EGFP
Vector sequence (SEQ ID NO: 193)
Plasmid 102: pLX-NLS_GFP-IRES-Hygro Vector sequence (SEQ ID NO: 194)
Plasmid 103: pLX-LifeAct_mCherry-IRES-Hygro
Vector sequence (SEQ ID NO: 195)
Plasmid 104: pLX-NLS_GFP-IRES-Hygro(l-89)-NpuDnaE(N)
Vector sequence (SEQ ID NO: 196)
Plasmid 105: pLX-LifeAct_mScarlet-IRES- NpuDnaE(C)-Hygro(90-341)
Vector sequence (SEQ ID NO: 197)
Plasmid 106: pX330-AAVS l
sgRNA spacer sequence: gACCCCACAGTGGGGCCACTA (First g does not match genome) (SEQ ID NO: 198) Vector sequence (SEQ ID NO: 199)
Plasmid 107: pAAVS l -Nst-EFl aHygro2ArtTA3(-)_TetO-Blast-P2A-EGFP
Vector sequence (SEQ ID NO:200)
Plasmid 108: pAAVS l -Nst-EFl aHygro2ArtTA3(-)_TetO-Blast-P2A-mScarlet
Vector sequence (SEQ ID NO:201)
Plasmid 109: pAAVS l -Nst-EFlaHygro2ArtTA3(-)_TetO-Blast(l-102)_NpuDnaE(N)-P2A-EGFP
Vector sequence (SEQ ID NO:202) Plasmid 110: pAAVS l -Nst-EFlaHygro2ArtTA3(-)_TetO-NpuDnaE(C)_Blast(103-140)-P2A-mScarlet Vector sequence (SEQ ID NO:203)
Plasmid 111: pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO-Hygro-P2A-NTR-E2A-EGFP
Vector sequence (SEQ ID NO:204)
Plasmid 112: pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO-Hygro-P2A-NTR-E2A-mCherry
Vector sequence (SEQ ID NO:205)
Plasmid 113: pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO- Hygro(l-89)-NpuDnaE(N)-P2A-NTR-E2A-EGFP Vector sequence (SEQ ID NO:206)
Plasmid 114: pAAVS l -Nst-EFl aBlast2ArtTA3(-)_TetO- NpuDnaE(C)-Hygro(90-341)-P2A-NTR-E2A-mCherry Vector sequence (SEQ ID NO:207) Plasmid 115: pLX-Hygro(l-89)_NpuDnaE(N)_LZA-IRES-TagBFP2
Protein = Hygro(l-89)-NpuDnaE(N)-LZA
Vector sequence (SEQ ID NO:208) Amino acid sequence (SEQ ID NO:209)
Plasmid 116: pLX-LZB_NpuDnaGEP(C)_Hygro(90-200)_SspDnaB(N-S0)-IRES-GFP
Protein = LZB-NpuDnaGEP(C)-Hygro(90-200)-SspDnaB(N-S0)
Vector sequence (SEQ ID NO:210)
Amino acid sequence (SEQ ID NO:211)
Plasmid 117: pLX-SspDnaB(C-S0)_Hygro(201-240)_NpuDnaE(N)_LZA-IRES-GFP
Protein = SspDnaB(C-S0)-Hygro(201-240)-NpuDnaE(N)-LZA
Vector sequence (SEQ ID NO:212)
Amino acid sequence (SEQ ID NO:213)
Plasmid 118: pLX-LZB_NpuDnaGEP(C)_Hygro(241-341)-IRES-mCherry
Protein = LZB-NpuDnaGEP(C)-Hygro(241-341)
Vector sequence (SEQ ID NO:214)
Amino acid sequence (SEQ ID NO:215)
AC1947GB (SEQ ID NO:216)
AC1949GB (SEQ ID NO:217)
pCR8-ccdbCam (SEQ ID NO:218)
References
I . Shearer, R.F. & Saunders, D.N. Experimental design for stable genetic manipulation in mammalian cell lines: lentivirus and alternatives. Genes to cells : devoted to molecular & cellular mechanisms 20, 1-10 (2015). 2. Abuin, A. & Bradley, A. Recycling selectable markers in mouse embryonic stem cells. Molecular and cellular biology 16, 1851-1856 (1996).
3. Shah, N.H. & Muir, T.W. Inteins: Nature's Gift to Protein Chemists. Chemical science 5, 446-461 (2014).
4. Zettler, J., Schtitz, V. & Mootz, H.D. The naturally split Npu DnaE intein exhibits an extraordinarily high rate in the protein trans-splicing reaction. FEBS letters 583, 909-914 (2009).
5. Iwai, H., Ztiger, S., Jin, J. & Tam, P.-H. Highly efficient protein trans-splicing by a naturally split DnaE intein from Nostoc punctiforme. FEBS letters 580, 1853-1858 (2006).
6. Sun, W., Yang, J. & Liu, X.Q. Synthetic two-piece and three-piece split inteins for protein trans-splicing. The Journal of biological chemistry 279, 35281-35286 (2004).
7. Cheriyan, M., Pedamallu, C.S., Tori, K. & Perler, F. Faster protein splicing with the Nostoc punctiforme DnaE intein using non-native extein residues. The Journal of biological chemistry 288, 6202-6211 (2013).
8. Chee, J. & Chin, C. Gateway cloning technology: Advantages and drawbacks. Cloning Transgenes 4, 138 (2015).
9. Bindels, D.S. et al. mScarlet: a bright monomeric red fluorescent protein for cellular imaging, nature methods
14, 53 (2017).
10. Stevens, A.J. et al. A promiscuous split intein with expanded protein engineering applications. Proceedings of the National Academy of Sciences 114, 8538-8543 (2017).
I I . Ghosh, I., Hamilton, A.D. & Regan, L. Antiparallel leucine zipper-directed protein reassembly: application to the green fluorescent protein. Journal of the American Chemical Society 122, 5658-5659 (2000).
12. Wang, H., La Russa, M. & Qi, L.S. CRISPR/Cas9 in genome editing and beyond. Annual review of
biochemistry 85, 227-264 (2016).
13. Peng, R., Lin, G. & Li, J. Potential pitfalls of CRISPR/Cas9-mediated genome editing. The FEBS journal 283,
1218-1231 (2016).
14. Oceguera-Yanez, F. et al. Engineering the AAVS 1 locus for consistent and scalable transgene expression in human iPSCs and their differentiated derivatives. Methods 101, 43-55 (2016). All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.
The indefinite articles "a" and "an," as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean "at least one."
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as
"comprising," "including," "carrying," "having," "containing," "involving," "holding," "composed of," and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases "consisting of and "consisting essentially of shall be closed or semi- closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
The terms "about" and "substantially" preceding a numerical value mean +10% of the recited numerical value.
Where a range of values is provided, each value between the upper and lower ends of the range are specifically contemplated and described herein.

Claims

What is claimed is: CLAIMS
1. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest; and
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the selectable marker protein to produce a full-length selectable marker protein.
2. The method of claim 1 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first and second vectors into the eukaryotic cells to produce transgenic eukaryotic cells.
3. The method of claim 2 further comprising selecting the transgenic eukaryotic cells that comprise the full-length selectable marker protein.
4. The method of any one of claims 1-3, wherein the eukaryotic cells are mammalian cells.
5. The method of any one of claims 1-4, wherein the selectable marker protein is an antibiotic resistance protein.
6. The method of claim 5, wherein the antibiotic resistance protein confers resistance to hygromycin, G418, puromycin, phleomycin Dl or blasticidin.
7. The method of claim 5 or 6, wherein the antibiotic resistance protein is encoded by a hygB gene, a bsr gene, a pac gene, or a neo gene.
8. The method of any one of claims 1-4, wherein the selectable marker protein is a fluorescent protein.
9. The method of claim 8, wherein the fluorescent protein is selected from TagCFP, mTagCFP2, Czurite, ECFP2, mKalamal, Sirius, Sapphire, T-Sapphire, ECFP, Cerulean, SCFP3C, mTurquoise, mTurquoise2, monomeric Midoriishi-Cyan, TagCFP, mTFPl, EGFP, Emerald, Superfolder GFP, Monomeric Czami Green, TagGFP2, mUKG, mWasabi, Clover, mNeonGreen, EYFP, Citrine, Venus, SYFP2, TagYFP, Monomeric Kusabira-Orange, ιηΚΟκ, mK02, mOrange, mOrange2, mRaspberry, mCherry, mStrawberry, mScarlet, mTangerine, tdTomato, TagRFP, TagRFP-T, mCpple, mRuby, mRuby2, mPlum, HcRed-Tandem, mKate2, mNeptune, NirFP, TagRFP657, IFP1.4 and iRFP.
10. The method of any one of claims 1-9, wherein the intein is a split intein.
11. The method of claim 10, wherein the split intein is a natural split, optionally wherein the natural split intein is selected from DnaE inteins, and optionally wherein the DnaE inteins are selected from Synechocystis sp. DnaE (SspOnaE) inteins and Nostoc punctiforme (TVpaDnaE) inteins.
12. The method of claim 10, wherein the split intein is an engineered split intein, optionally wherein the engineered split intein is engineered from DnaB inteins or GyrB inteins, and optionally wherein the engineered split intein is a SspOna S 1 intein or a SspGyrB S 11 intein.
13. The method of any one of claims 1-12, wherein the first and/or second molecule is a protein or a non-coding ribonucleic acid (RNA), optionally wherein the non-coding RNA is a microRNA (miRNA), antisense RNA, short-interfering RNA (siRNA) or short-hairpin RNA (shRNA).
14. The method of any one of claims 1-13, wherein the first and/or second vector is a plasmid vector or a viral vector.
15. A eukaryotic cell, comprising
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein, which is upstream from a nucleotide sequence encoding an N- terminal fragment of an intein and (ii) a nucleotide sequence encoding a first molecule of interest; and
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a second molecule of interest,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the selectable marker protein to produce a full-length antibiotic resistance protein.
16. A kit, comprising
(a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of an intein; and
(b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the intein, which is upstream from a C-terminal fragment of the selectable marker protein,
wherein the N-terminal fragment and the C-terminal fragment of the intein catalyze joining of the N-terminal fragment and the C-terminal fragment of the selectable marker protein to produce a full-length selectable marker protein.
17. A method comprising delivering to eukaryotic cells
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the central fragment of the selectable marker protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the selectable marker protein to the C-terminal fragment of the selectable marker protein, to produce a full-length selectable marker protein.
18. The method of claim 19 further comprising maintaining the eukaryotic cells under conditions that permit introduction of the first, second, and third vectors into the eukaryotic cells to produce transgenic eukaryotic
19. The method of claim 18 further comprising selecting the transgenic eukaryotic cells that comprise the full-length selectable marker protein.
20. A eukaryotic cell comprising:
(a) a first vector comprising (i) a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein and (ii) a nucleotide sequence encoding a first molecule of interest,
(b) a second vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein and (ii) a nucleotide sequence encoding a second molecule of interest, and
(c) a third vector comprising (i) a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein and (ii) a nucleotide sequence encoding a third molecule of interest, wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the central fragment of the selectable marker protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the selectable marker protein to the C-terminal fragment of the selectable marker protein, to produce a full-length selectable marker protein.
21. A kit comprising:
(a) a first vector comprising a nucleotide sequence encoding an N-terminal fragment of a selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a first intein,
(b) a second vector comprising a nucleotide sequence encoding a C-terminal fragment of the first intein, which is upstream from a nucleotide sequence encoding a central fragment of the selectable marker protein, which is upstream from a nucleotide sequence encoding an N-terminal fragment of a second intein, and
(c) a third vector comprising a nucleotide sequence encoding a C-terminal fragment of the second intein, which is upstream from a nucleotide sequence encoding a C-terminal fragment of the selectable marker protein,
wherein the N-terminal fragment and the C-terminal fragment of the first intein catalyze joining of N-terminal fragment of the selectable marker protein to the central fragment of the selectable marker protein, and the N-terminal fragment and the C-terminal fragment of the second intein catalyze joining of central fragment of the selectable marker protein to the C-terminal fragment of the selectable marker protein, to produce a full-length selectable marker protein.
PCT/US2018/055412 2017-10-12 2018-10-11 Transgenic selection methods and compositions WO2019075200A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA3079017A CA3079017A1 (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions
US16/755,065 US20200263197A1 (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions
CN201880078542.7A CN111511759A (en) 2017-10-12 2018-10-11 Transgene selection methods and compositions
JP2020520468A JP7394752B2 (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions
AU2018347421A AU2018347421A1 (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions
EP18867279.4A EP3694869A4 (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions
KR1020207013411A KR20200064129A (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions
JP2023200808A JP2024015079A (en) 2017-10-12 2023-11-28 Transgenic selection methods and compositions

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201762571672P 2017-10-12 2017-10-12
US62/571,672 2017-10-12
US201762608478P 2017-12-20 2017-12-20
US62/608,478 2017-12-20
US201862616281P 2018-01-11 2018-01-11
US62/616,281 2018-01-11
US201862624629P 2018-01-31 2018-01-31
US62/624,629 2018-01-31

Publications (1)

Publication Number Publication Date
WO2019075200A1 true WO2019075200A1 (en) 2019-04-18

Family

ID=66101179

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/055412 WO2019075200A1 (en) 2017-10-12 2018-10-11 Transgenic selection methods and compositions

Country Status (8)

Country Link
US (1) US20200263197A1 (en)
EP (1) EP3694869A4 (en)
JP (2) JP7394752B2 (en)
KR (1) KR20200064129A (en)
CN (1) CN111511759A (en)
AU (1) AU2018347421A1 (en)
CA (1) CA3079017A1 (en)
WO (1) WO2019075200A1 (en)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11293021B1 (en) 2016-06-23 2022-04-05 Inscripta, Inc. Automated cell processing methods, modules, instruments, and systems
US9982279B1 (en) 2017-06-23 2018-05-29 Inscripta, Inc. Nucleic acid-guided nucleases
US10011849B1 (en) 2017-06-23 2018-07-03 Inscripta, Inc. Nucleic acid-guided nucleases
JP2020530979A (en) 2017-06-30 2020-11-05 インスクリプタ, インコーポレイテッド Automatic cell processing methods, modules, equipment and systems
US10557216B2 (en) 2018-04-24 2020-02-11 Inscripta, Inc. Automated instrumentation for production of T-cell receptor peptide libraries
US10858761B2 (en) 2018-04-24 2020-12-08 Inscripta, Inc. Nucleic acid-guided editing of exogenous polynucleotides in heterologous cells
AU2019292919A1 (en) 2018-06-30 2021-03-11 Inscripta, Inc. Instruments, modules, and methods for improved detection of edited sequences in live cells
US11142740B2 (en) 2018-08-14 2021-10-12 Inscripta, Inc. Detection of nuclease edited sequences in automated modules and instruments
US11214781B2 (en) 2018-10-22 2022-01-04 Inscripta, Inc. Engineered enzyme
CN113631713A (en) 2019-03-25 2021-11-09 因思科瑞普特公司 Simultaneous multiplex genome editing in yeast
US11001831B2 (en) 2019-03-25 2021-05-11 Inscripta, Inc. Simultaneous multiplex genome editing in yeast
CN113939593A (en) 2019-06-06 2022-01-14 因思科瑞普特公司 Treatment for recursive nucleic acid-guided cell editing
WO2021102059A1 (en) 2019-11-19 2021-05-27 Inscripta, Inc. Methods for increasing observed editing in bacteria
AU2020407048A1 (en) 2019-12-18 2022-06-09 Inscripta, Inc. Cascade/dCas3 complementation assays for in vivo detection of nucleic acid-guided nuclease edited cells
CN115135370A (en) 2020-01-27 2022-09-30 因思科瑞普特公司 Electroporation module and instrument
US20210332388A1 (en) 2020-04-24 2021-10-28 Inscripta, Inc. Compositions, methods, modules and instruments for automated nucleic acid-guided nuclease editing in mammalian cells
US11787841B2 (en) 2020-05-19 2023-10-17 Inscripta, Inc. Rationally-designed mutations to the thrA gene for enhanced lysine production in E. coli
WO2022060749A1 (en) 2020-09-15 2022-03-24 Inscripta, Inc. Crispr editing to embed nucleic acid landing pads into genomes of live cells
US11512297B2 (en) 2020-11-09 2022-11-29 Inscripta, Inc. Affinity tag for recombination protein recruitment
WO2022146497A1 (en) 2021-01-04 2022-07-07 Inscripta, Inc. Mad nucleases
WO2022150269A1 (en) 2021-01-07 2022-07-14 Inscripta, Inc. Mad nucleases
US11884924B2 (en) 2021-02-16 2024-01-30 Inscripta, Inc. Dual strand nucleic acid-guided nickase editing
EP4301861A1 (en) 2021-03-03 2024-01-10 Shape Therapeutics Inc. Auxotrophic cells for virus production and compositions and methods of making
WO2023027169A1 (en) * 2021-08-27 2023-03-02 国立大学法人 東京医科歯科大学 Sorting system for live cells
WO2023027170A1 (en) * 2021-08-27 2023-03-02 国立大学法人 東京医科歯科大学 System for regulating protein translation
CN115896147B (en) * 2022-10-11 2023-10-03 态创生物科技(广州)有限公司 Intein evolution systems and methods, corresponding mutant plasmids and reporter plasmids

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6858775B1 (en) * 1999-05-24 2005-02-22 New England Biolabs, Inc. Method for generating split, non-transferable genes that are able to express an active protein product
US20050221343A1 (en) * 2003-10-24 2005-10-06 Waldo Geoffrey S Self-assembling split-fluorescent protein systems
US20070065912A1 (en) * 2005-07-21 2007-03-22 Abbott Laboratories Multiple Gene Expression including sORF Constructs and Methods with Polyproteins, Pro-Proteins, and Proteolysis
US20150232507A1 (en) * 2011-09-28 2015-08-20 Era Biotech, S.A. Split inteins and uses thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH10113174A (en) * 1996-10-08 1998-05-06 Amashiyamu Kk Simultaneous production of human cytochrome p-450 and human cytochrome p450 reductase
DE60036942T2 (en) * 1999-05-24 2008-08-07 New England Biolabs, Inc., Beverly METHOD OF GENERATING SEPARATE, NON-TRANSFERABLE GENES WHICH ARE ABLE TO EXPRESS AN ACTIVE PROTEIN PRODUCT
CN101993907B (en) * 2002-01-08 2018-12-28 迈克尔·R·拉尔布 Expressing CIVPS or the genetically modified plants of intein modified proteins and preparation method thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6858775B1 (en) * 1999-05-24 2005-02-22 New England Biolabs, Inc. Method for generating split, non-transferable genes that are able to express an active protein product
US20050221343A1 (en) * 2003-10-24 2005-10-06 Waldo Geoffrey S Self-assembling split-fluorescent protein systems
US20070065912A1 (en) * 2005-07-21 2007-03-22 Abbott Laboratories Multiple Gene Expression including sORF Constructs and Methods with Polyproteins, Pro-Proteins, and Proteolysis
US20150232507A1 (en) * 2011-09-28 2015-08-20 Era Biotech, S.A. Split inteins and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JILLETTE ET AL.: "Split Selectable Markers", BIORXIV, 25 October 2018 (2018-10-25), pages 1 - 27, XP055592242 *
See also references of EP3694869A4 *
VOLKMANN ET AL.: "Protein trans-splicing and its use in structural biology: opportunities and limitations", MOLECULAR BIOSYSTEMS, vol. 6, no. 11, 31 August 2010 (2010-08-31), pages 2110 - 2121, XP055592232 *

Also Published As

Publication number Publication date
JP7394752B2 (en) 2023-12-08
JP2024015079A (en) 2024-02-01
KR20200064129A (en) 2020-06-05
CA3079017A1 (en) 2019-04-18
AU2018347421A1 (en) 2020-05-14
US20200263197A1 (en) 2020-08-20
EP3694869A4 (en) 2021-11-24
EP3694869A1 (en) 2020-08-19
CN111511759A (en) 2020-08-07
JP2020537646A (en) 2020-12-24

Similar Documents

Publication Publication Date Title
JP7394752B2 (en) Transgenic selection methods and compositions
US10344285B2 (en) DNA vectors, transposons and transposases for eukaryotic genome modification
US10435696B2 (en) DNA vectors, transposons and transposases for eukaryotic genome modification
Jillette et al. Split selectable markers
WO2019016175A9 (en) A two-component vector library system for rapid assembly and diversification of full-length t-cell receptor open reading frames
US20200087679A1 (en) Expression cassette
TW200930815A (en) Novel recombination sequences
US20220195465A1 (en) Stable targeted integration
JP5246904B2 (en) Vector for introducing foreign gene and method for producing vector into which foreign gene has been introduced
JP7472167B2 (en) Stable targeted integration
EP4311858A1 (en) Intein-based controllers
WO2023064871A1 (en) Integrases, landing pad architectures, and engineered cells comprising the same
WO2024068995A1 (en) Novel transposase system
WO2023232961A1 (en) Improved production cells
CN117355607A (en) Non-viral homology mediated end ligation
CA3143506A1 (en) Enhanced platforms for unnatural amino acid incorporation in mammalian cells
Toktay Engineering and systematic comparison of constitutive promoters in various lines of chinese hamster ovary cells
NZ626252B2 (en) Expression cassette

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18867279

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3079017

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020520468

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20207013411

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018347421

Country of ref document: AU

Date of ref document: 20181011

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018867279

Country of ref document: EP

Effective date: 20200512